1
|
Reisdorph R, Littrell-Miller B, Powell R, Reisdorph N. A mass spectrometry based predictive strategy reveals ADAP1 is phosphorylated at tyrosine 364. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2018; 32:1173-1180. [PMID: 29659066 DOI: 10.1002/rcm.8140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/26/2018] [Accepted: 03/30/2018] [Indexed: 06/08/2023]
Abstract
RATIONALE The goal of this work was to identify phosphorylation sites within the amino acid sequence of human ADAP1. Using traditional mass spectrometry based techniques we were unable to produce interpretable spectra demonstrating modification by phosphorylation. This prompted us to employ a strategy in which phosphorylated peptides were first predicted using peptide mapping followed by targeted MS/MS acquisition. METHODS ADAP1 was immunoprecipitated from extracts of HEK293 cells stably transfected with ADAP1 cDNA. Immunoprecipitated ADAP1 was digested with proteolytic enzymes and analyzed by LC/MS in MS1 mode by high-resolution quadrupole time-of-flight mass spectrometry (QTOF-MS). Peptide molecular features were extracted using an untargeted data-mining algorithm. Extracted peptide neutral masses were matched against the ADAP1 amino acid sequence with phosphorylation included as a predicted modification. Peptides with predicted phosphorylation sites were analyzed by targeted LC/MS2 . Acquired MS2 spectra were then analyzed using database search engines to confirm phosphorylation. Spectra of phosphorylated peptides were validated by manual interpretation. Further confirmation was performed by manipulating phospho-peptide abundance using calf intestinal phosphatase (CIP) and the phorbol ester, phorbol 12-myristate 13-acetate (PMA). RESULTS Of five predicted phosphopeptides, one, comprised of the sequence AVDRPMLPQEYAVEAHFK, was confirmed to be phosphorylated on a tyrosine at position 364. Pre-treatment of cells with PMA prior to immunoprecipitation increased the ratio of phosphorylated to unphosphorylated peptide as determined by area counts of extracted ion chromatograms (EIC). Addition of CIP to immunoprecipitation reactions eliminated the phosphorylated form. CONCLUSIONS A novel phosphorylation site was identified at tyrosine 364. Phosphorylation at this site is increased by treatment with PMA. PMA promotes membrane translocation and activation of protein kinase C (PKC), indicating that tyrosine 364 is phosphorylated by a PKC-dependent mechanism.
Collapse
Affiliation(s)
- Richard Reisdorph
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - BobbiJo Littrell-Miller
- Environment, Safety, Health & Quality Office, National Renewable Energy Laboratory (NREL), 15013 Denver West Parkway, Golden, CO, USA
| | - Roger Powell
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Nichole Reisdorph
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
2
|
ADAP2 Is an Interferon Stimulated Gene That Restricts RNA Virus Entry. PLoS Pathog 2015; 11:e1005150. [PMID: 26372645 PMCID: PMC4570769 DOI: 10.1371/journal.ppat.1005150] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/13/2015] [Indexed: 12/19/2022] Open
Abstract
Interferon stimulated genes (ISGs) target viruses at various stages of their infectious life cycles, including at the earliest stage of viral entry. Here we identify ArfGAP with dual pleckstrin homology (PH) domains 2 (ADAP2) as a gene upregulated by type I IFN treatment in a STAT1-dependent manner. ADAP2 functions as a GTPase-activating protein (GAP) for Arf6 and binds to phosphatidylinositol 3,4,5-trisphosphate (PI(3,4,5)P3) and PI(3,4)P2. We show that overexpression of ADAP2 suppresses dengue virus (DENV) and vesicular stomatitis virus (VSV) infection in an Arf6 GAP activity-dependent manner, while exerting no effect on coxsackievirus B (CVB) or Sendai virus (SeV) replication. We further show that ADAP2 expression induces macropinocytosis and that ADAP2 strongly associates with actin-enriched membrane ruffles and with Rab8a- and LAMP1-, but not EEA1- or Rab7-, positive vesicles. Utilizing two techniques--light-sensitive neutral red (NR)-containing DENV and fluorescence assays for virus internalization--we show that ADAP2 primarily restricts DENV infection at the stage of virion entry and/or intracellular trafficking and that incoming DENV and VSV particles associate with ADAP2 during their entry. Taken together, this study identifies ADAP2 as an ISG that exerts antiviral effects against RNA viruses by altering Arf6-mediated trafficking to disrupt viral entry.
Collapse
|
3
|
Durinck S, Stawiski EW, Pavía-Jiménez A, Modrusan Z, Kapur P, Jaiswal BS, Zhang N, Toffessi-Tcheuyap V, Nguyen TT, Pahuja KB, Chen YJ, Saleem S, Chaudhuri S, Heldens S, Jackson M, Peña-Llopis S, Guillory J, Toy K, Ha C, Harris CJ, Holloman E, Hill HM, Stinson J, Rivers CS, Janakiraman V, Wang W, Kinch LN, Grishin NV, Haverty PM, Chow B, Gehring JS, Reeder J, Pau G, Wu TD, Margulis V, Lotan Y, Sagalowsky A, Pedrosa I, de Sauvage FJ, Brugarolas J, Seshagiri S. Spectrum of diverse genomic alterations define non-clear cell renal carcinoma subtypes. Nat Genet 2014; 47:13-21. [PMID: 25401301 DOI: 10.1038/ng.3146] [Citation(s) in RCA: 279] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 10/24/2014] [Indexed: 12/17/2022]
Abstract
To further understand the molecular distinctions between kidney cancer subtypes, we analyzed exome, transcriptome and copy number alteration data from 167 primary human tumors that included renal oncocytomas and non-clear cell renal cell carcinomas (nccRCCs), consisting of papillary (pRCC), chromophobe (chRCC) and translocation (tRCC) subtypes. We identified ten significantly mutated genes in pRCC, including MET, NF2, SLC5A3, PNKD and CPQ. MET mutations occurred in 15% (10/65) of pRCC samples and included previously unreported recurrent activating mutations. In chRCC, we found TP53, PTEN, FAAH2, PDHB, PDXDC1 and ZNF765 to be significantly mutated. Gene expression analysis identified a five-gene set that enabled the molecular classification of chRCC, renal oncocytoma and pRCC. Using RNA sequencing, we identified previously unreported gene fusions, including ACTG1-MITF fusion. Ectopic expression of the ACTG1-MITF fusion led to cellular transformation and induced the expression of downstream target genes. Finally, we observed upregulation of the anti-apoptotic factor BIRC7 in MiTF-high RCC tumors, suggesting a potential therapeutic role for BIRC7 inhibitors.
Collapse
Affiliation(s)
- Steffen Durinck
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA.,Bioinformatics and Computational Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Eric W Stawiski
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA.,Bioinformatics and Computational Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Andrea Pavía-Jiménez
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zora Modrusan
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Payal Kapur
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bijay S Jaiswal
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Na Zhang
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Vanina Toffessi-Tcheuyap
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thong T Nguyen
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Kanika Bajaj Pahuja
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Ying-Jiun Chen
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Sadia Saleem
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Subhra Chaudhuri
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Sherry Heldens
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Marlena Jackson
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Samuel Peña-Llopis
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph Guillory
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Karen Toy
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Connie Ha
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Corissa J Harris
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Eboni Holloman
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Haley M Hill
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jeremy Stinson
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | | | | | - Weiru Wang
- Structural Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Lisa N Kinch
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Nick V Grishin
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Peter M Haverty
- Bioinformatics and Computational Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Bernard Chow
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Julian S Gehring
- Bioinformatics and Computational Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Jens Reeder
- Bioinformatics and Computational Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Gregoire Pau
- Bioinformatics and Computational Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Thomas D Wu
- Bioinformatics and Computational Biology Department, Genentech, Inc., South San Francisco, California, USA
| | - Vitaly Margulis
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yair Lotan
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Arthur Sagalowsky
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ivan Pedrosa
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Frederic J de Sauvage
- Molecular Oncology Department, Genentech, Inc., South San Francisco, California, USA
| | - James Brugarolas
- Kidney Cancer Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Somasekar Seshagiri
- Molecular Biology Department, Genentech, Inc., South San Francisco, California, USA
| |
Collapse
|
4
|
Identification of components of the host type IA phosphoinositide 3-kinase pathway that promote internalization of Listeria monocytogenes. Infect Immun 2011; 80:1252-66. [PMID: 22158742 DOI: 10.1128/iai.06082-11] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The bacterial pathogen Listeria monocytogenes causes food-borne illnesses resulting in gastroenteritis, meningitis, or abortion. Listeria promotes its internalization into some human cells through binding of the bacterial surface protein InlB to the host receptor tyrosine kinase Met. The interaction of InlB with the Met receptor stimulates host signaling pathways that promote cell surface changes driving bacterial uptake. One human signaling protein that plays a critical role in Listeria entry is type IA phosphoinositide 3-kinase (PI 3-kinase). The molecular mechanism by which PI 3-kinase promotes bacterial internalization is not understood. Here we perform an RNA interference (RNAi)-based screen to identify components of the type IA PI 3-kinase pathway that control the entry of Listeria into the human cell line HeLa. The 64 genes targeted encode known upstream regulators or downstream effectors of type IA PI 3-kinase. The results of this screen indicate that at least 9 members of the PI 3-kinase pathway play important roles in Listeria uptake. These 9 human proteins include a Rab5 GTPase, several regulators of Arf or Rac1 GTPases, and the serine/threonine kinases phosphoinositide-dependent kinase 1 (PDK1), mammalian target of rapamycin (mTor), and protein kinase C-ζ. These findings represent a key first step toward understanding the mechanism by which type IA PI 3-kinase controls bacterial internalization.
Collapse
|
5
|
Wang H, Ma J, Ruan L, Xu X. Cloning of a centaurin-alpha1 like gene MjCent involved in WSSV infection from shrimp Marsupeneaus japonicus. FISH & SHELLFISH IMMUNOLOGY 2009; 26:279-284. [PMID: 19073266 DOI: 10.1016/j.fsi.2008.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2008] [Revised: 10/22/2008] [Accepted: 10/28/2008] [Indexed: 05/27/2023]
Abstract
Centaurin-alpha1 specifically binds phosphatidylinositol 3,4,5-trisphosphate (PI (3,4,5)P3) and is a GTPase-activating protein (GAP) of ADP-ribosylation factor (ARF6). It actively engages in phosphatidylinositol 3-kinase (PI3-K) mediated cell signal transduction. Here, for the first time, we have identified a virus related centaurin-alpha1 homologue named MjCent from the shrimp, Marsupeneaus japonicus, an economically important crustacean in the aquaculture industry. MjCent has one conserved ArfGAP and two Pleckstrin homology domains (PH domains). As shown by RT-PCR and immunofluorescence, MjCent appeared in every tissue examined and was localized mainly in the cell cytoplasm. Further investigation with real-time quantitative PCR showed that MjCent was significantly up-regulated during white spot syndrome virus (WSSV) infection, but notably decreased in virus-resistant shrimps. This suggests a close relationship between MjCent and WSSV invasion and host defense of the shrimp, M. japonicus.
Collapse
Affiliation(s)
- Huifen Wang
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, State Oceanic Administration, Xiamen, 361005, PR China
| | | | | | | |
Collapse
|
6
|
Venkateswarlu K, Brandom KG, Yun H. PI-3-kinase-dependent membrane recruitment of centaurin-alpha2 is essential for its effect on ARF6-mediated actin cytoskeleton reorganisation. J Cell Sci 2007; 120:792-801. [PMID: 17284522 DOI: 10.1242/jcs.03373] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
GTPase activating proteins (GAPs) of the centaurin family regulate the actin cytoskeleton and vesicle trafficking through inactivation of the ADP-ribosylation factor (ARF) family of small GTP-binding proteins. We report the functional characterisation of centaurin-alpha(2), which is structurally related to the centaurin-alpha(1) ARF6 GAP. centaurin-alpha(2) contains an N-terminal GAP domain followed by two pleckstrin homology (PH) domains (N-PH and C-PH). In vitro, GFP-centaurin-alpha(2) specifically binds the phosphatidylinositol (PI) 3-kinase lipid products, PI 3,4-P(2) and PI 3,4,5-P(3) (PIP(3)), through its C-terminal PH domain. In agreement with this observation, GFP-centaurin-alpha(2) was recruited to the plasma membrane from the cytosol in EGF-stimulated cells in a PI-3-kinase-dependent manner. Moreover, the C-PH domain is sufficient and necessary for membrane recruitment of centaurin-alpha(2). centaurin-alpha(2) shows sustained kinetics of PI-3-kinase-mediated membrane recruitment in EGF-stimulated cells, owing to its binding to PI 3,4-P(2). centaurin-alpha(2) prevents ARF6 translocation to, and cortical actin formation at, the plasma membrane, which are phenotypic indications for ARF6 activation in EGF-stimulated cells. Moreover, the constitutively active mutant of ARF6 reverses the effect of centaurin-alpha(2) on cortical actin formation. The membrane targeted centaurin-alpha(2) is constitutively active. Together, these studies indicate that centaurin-alpha(2) is recruited in a sustained manner to the plasma membrane through binding to PI 3,4-P(2) and thereby regulates actin reorganisation via ARF6.
Collapse
|
7
|
Abstract
Glutamate receptors regulate gene expression in neurons by activating intracellular signaling cascades that phosphorylate transcription factors within the nucleus. The mitogen-activated protein kinase (MAPK) cascade is one of the best characterized cascades in this regulatory process. The Ca(2+)-permeable ionotropic glutamate receptor, mainly the NMDA receptor subtype, activates MAPKs through a biochemical route involving the Ca(2+)-sensitive Ras-guanine nucleotide releasing factor, Ca(2+)/calmodulin-dependent protein kinase II, and phosphoinositide 3-kinase. The metabotropic glutamate receptor (mGluR), however, activates MAPKs primarily through a Ca(2+)-insensitve pathway involving the transactivation of receptor tyrosine kinases. The adaptor protein Homer also plays a role in this process. As an information superhighway between surface glutamate receptors and transcription factors in the nucleus, active MAPKs phosphorylate specific transcription factors (Elk-1 and CREB), and thereby regulate distinct programs of gene expression. The regulated gene expression contributes to the development of multiple forms of synaptic plasticity related to long-lasting changes in memory function and addictive properties of drugs of abuse. This review, by focusing on new data from recent years, discusses the signaling mechanisms by which different types of glutamate receptors activate MAPKs, features of each MAPK cascade in regulating gene expression, and the importance of glutamate/MAPK-dependent synaptic plasticity in memory and addiction.
Collapse
Affiliation(s)
- John Q Wang
- Department of Basic Medical Science, University of Missouri-Kansas City, School of Medicine, Kansas City, Missouri, USA.
| | | | | |
Collapse
|
8
|
Centaurin-alpha1 and KIF13B kinesin motor protein interaction in ARF6 signalling. Biochem Soc Trans 2005; 33:1279-81. [PMID: 16246098 DOI: 10.1042/bst0331279] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The ARF (ADP-ribosylation factor) family of small GTPases regulate intracellular membrane trafficking by cycling between an inactive GDP- and an active GTP-bound form. Among the six known mammalian ARFs (ARF1-ARF6), ARF6 is the least conserved and plays critical roles in membrane trafficking and cytoskeletal dynamics near the cell surface. Since ARFs have undetectable levels of intrinsic GTP binding and hydrolysis, they are totally dependent on extrinsic GEFs (guanine nucleotide-exchange factors) for GTP binding and GAPs (GTPase-activating proteins) for GTP hydrolysis. We have recently isolated a novel KIF (kinesin) motor protein (KIF13B) that binds to centaurin-alpha1, an ARF6GAP that binds to the second messenger PIP3 [PtdIns(3,4,5)P3]. KIFs transport intracellular vesicles and recognize their cargo by binding to proteins (receptors) localized on the surface of the cargo vesicles. Identification of centaurin-alpha1 as a KIF13B interactor suggests that KIF13B may transport ARF6 and/or PIP3 using centaurin-alpha1 as its receptor. This paper reviews the studies carried out to assess the interaction and regulation of centaurin-alpha1 by KIF13B.
Collapse
|
9
|
Hayashi H, Matsuzaki O, Muramatsu S, Tsuchiya Y, Harada T, Suzuki Y, Sugano S, Matsuda A, Nishida E. Centaurin-alpha1 is a phosphatidylinositol 3-kinase-dependent activator of ERK1/2 mitogen-activated protein kinases. J Biol Chem 2005; 281:1332-7. [PMID: 16287813 DOI: 10.1074/jbc.m505905200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Centaurin-alpha1 is known to be a phosphatidylinositol 3,4,5-triphosphate (PIP3)-binding protein that has two pleckstrin homology domains and a putative ADP ribosylation factor GTPase-activating protein domain. However, the physiological function of centaurin-alpha1 is still not understood. Here we have shown that transient expression of centaurin-alpha1 in COS-7 cells results in specific activation of ERK, and the activation is inhibited by co-expression of a dominant negative form of Ras. We have also found that a mutant form of centaurin-alpha1 that is unable to bind PIP3 fails to induce ERK activation and that a phosphatidylinositol 3-kinase inhibitor LY294002 inhibits centaurin-alpha1-dependent ERK activation. Furthermore, transient knockdown of centaurin-alpha1 by small interfering RNAs results in reduced ERK activation after epidermal growth factor stimulation in T-REx 293 cells. These results suggest that centaurin-alpha1 contributes to ERK activation in growth factor signaling, linking the PI3K pathway to the ERK mitogen-activated protein kinase pathway through its ability to interact with PIP3.
Collapse
Affiliation(s)
- Hideko Hayashi
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Venkateswarlu K, Hanada T, Chishti AH. Centaurin-α1 interacts directly with kinesin motor protein KIF13B. J Cell Sci 2005; 118:2471-84. [PMID: 15923660 DOI: 10.1242/jcs.02369] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Centaurin-α1 is a phosphatidylinositol 3,4,5-trisphosphate binding protein as well as a GTPase activating protein (GAP) for the ADP-ribosylation factor (ARF) family of small GTPases. To further understand its cellular function, we screened a rat brain cDNA library using centaurin-α1 as bait to identify centaurin-α1 interacting proteins. The yeast two-hybrid screen identified a novel kinesin motor protein as a centaurin-α1 binding partner. The motor protein, termed KIF13B, encoded by a single ∼9.5-kb transcript, is widely expressed with high levels observed in brain and kidney. Yeast two-hybrid and GST pull-down assays showed that the interaction between centaurin-α1 and KIF13B is direct and mediated by the GAP domain of centaurin-α1 and the stalk domain of KIF13B. Centaurin-α1 and KIF13B form a complex in vivo and the KIF13B interaction appears to be specific to centaurin-α1 as other members of the ARF GAP family did not show any binding activity. We also show that KIF13B and centaurin-α1 colocalize at the leading edges of the cell periphery whereas a deletion mutant of centaurin-α1 that lacks the KIF13B binding site, failed to colocalize with KIF13B in vivo. Finally, we demonstrate that KIF13B binding suppresses the ARF6 GAP activity of centaurin-α1 in intact cells. Together, our data suggest a mechanism where direct binding between centaurin-α1 and KIF13B could concentrate centaurin-α1 at the leading edges of cells, thus modulating ARF6 function.
Collapse
Affiliation(s)
- Kanamarlapudi Venkateswarlu
- Department of Pharmacology, School of Medical Sciences, The University of Bristol, University Walk, Bristol, BS8 1TD, UK.
| | | | | |
Collapse
|
11
|
Lawrence J, Mundell SJ, Yun H, Kelly E, Venkateswarlu K. Centaurin-α1, an ADP-Ribosylation Factor 6 GTPase Activating Protein, Inhibits β2-Adrenoceptor Internalization. Mol Pharmacol 2005; 67:1822-8. [PMID: 15778454 DOI: 10.1124/mol.105.011338] [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: 11/22/2022] Open
Abstract
The small GTP-binding protein ADP ribosylation factor 6 (ARF6) has recently been implicated in the internalization of G protein-coupled receptors (GPCRs), although its precise molecular mechanism in this process remains unclear. We have recently identified centaurin alpha(1) as a GTPase activating protein (GAP) for ARF6. In the current study, we characterized the effects of centaurin alpha(1) on the agonist-induced internalization of the beta(2)-adrenoceptor transiently expressed in human embryonic kidney (HEK) 293 cells. Using an enzyme-linked immunosorbent assay as well as confocal imaging of cells, we found that expression of centaurin alpha(1) strongly inhibited the isoproterenol-induced internalization of beta(2)-adrenoceptor. On the other hand, expression of functionally inactive versions of centaurin alpha(1), including an R49C mutant, which has no catalytic activity, and a double pleckstrin homology (PH) mutant (DM; R148C/R273C), which has mutations in both the PH domains of centaurin alpha(1), rendering it unable to translocate to the cell membrane, were unable to inhibit beta(2)-adrenoceptor internalization. In addition, a constitutively active version of ARF6, ARF6Q67L, reversed the ability of centaurin alpha(1) to inhibit beta(2)-adrenoceptor internalization. Finally, expression of centaurin alpha(1) also inhibited the agonist-induced internalization of beta(2)-adrenoceptor endogenously expressed in HEK 293 cells, whereas the R49C and DM mutant versions of centaurin alpha(1) had no effect. Together, these data indicate that by acting as an ARF6 GAP, centaurin alpha(1) is able to switch off ARF6 and so inhibit its ability to mediate beta(2)-adrenoceptor internalization. Thus, ARF6 GAPs, such as centaurin alpha(1), are likely to play a crucial role in GPCR trafficking by modulating the activity of ARF6.
Collapse
Affiliation(s)
- Joanna Lawrence
- Department of Pharmacology, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | | | | | | | | |
Collapse
|
12
|
Reiser G, Bernstein HG. Altered expression of protein p42IP4/centaurin-alpha 1 in Alzheimer's disease brains and possible interaction of p42IP4 with nucleolin. Neuroreport 2004; 15:147-8. [PMID: 15106847 DOI: 10.1097/00001756-200401190-00028] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The protein p42IP4 (centaurin-alpha1) is a brain-specific InsP4/PtdIns3 (PIP)-binding protein, whish is localized in neurons of the human brain. In Alzheimer's disease (AD) the intraneuronal expression of the protein was shown to be elevated. In addition, p42IP4 immunostaining decorated neuritic plaques. Attempting to explain the putative role of the protein in AD, we have concentrated on its well-known interactions with casein kinase I, which is known to be prominently involved in AD pathophysiology. Meanwhile, specific interaction of p42IP4 with nucleolin, another player in AD pathology, has been revealed. Based on these data, we propose alternative concepts of how p42IP4 might act in AD pathomechanisms.
Collapse
Affiliation(s)
- Georg Reiser
- Institut für Neurobiochemie, Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Germany.
| | | |
Collapse
|
13
|
Thacker E, Kearns B, Chapman C, Hammond J, Howell A, Theibert A. The Arf6 GAP centaurin α-1 is a neuronal actin-binding protein which also functions via GAP-independent activity to regulate the actin cytoskeleton. Eur J Cell Biol 2004; 83:541-54. [PMID: 15679100 DOI: 10.1078/0171-9335-00416] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Centaurin alpha-1 is a high-affinity PtdIns(3,4,5)P3-binding protein enriched in brain. Sequence analysis indicates centaurin alpha-1 contains two pleckstrin homology domains, ankyrin repeats and an Arf GAP homology domain, placing it in the AZAP family of phosphoinositide-regulated Arf GAPs. Other members of this family are involved in actin cytoskeletal and focal adhesion organization. Recently, it was reported that centaurin alpha-1 expression diminishes cortical actin and decreases Arf6GTP levels consistent with it functioning as an Arf6 GAP in vivo. In the current report, we show that centaurin alpha-1 binds Arfs in vitro and colocalizes with Arf6 and Arf5 in vivo, further supporting an interaction with Arfs. Centaurin alpha-1 expression produces dramatic effects on the actin cytoskeleton, decreasing stress fibers, diminishing cortical actin, and enhancing membrane ruffles and filopodia. Expression of centaurin alpha-1 also enhances cell spreading and disrupts focal adhesion protein localization. The effects of centaurin alpha-1 on stress fibers and cell spreading are reminiscent of those of Arf6GTP. Consistent with this, we show that many of the centaurin alpha-1-induced effects on the actin cytoskeleton and actin-dependent activities do not require GAP activity. Thus, centaurin alpha-1 likely functions via both GAP-dependent and GAP-independent mechanisms to regulate the actin cytoskeleton. Furthermore, we demonstrate that in vitro, centaurin alpha-1 binds F-actin directly, with actin binding activity localized to the PtdIns(3,4,5)P3-binding PH domain. Our data suggest that centaurin alpha-1 may be a component of the neuronal PI 3-kinase cascade that leads to regulation of the neuronal actin cytoskeleton.
Collapse
Affiliation(s)
- Erin Thacker
- Department of Neurobiology and Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
14
|
Hanck T, Stricker R, Sedehizade F, Reiser G. Identification of gene structure and subcellular localization of human centaurin α2, and p42IP4, a family of two highly homologueous, Ins 1,3,4,5-P4-/PtdIns 3,4,5-P3-binding, adapter proteins. J Neurochem 2003; 88:326-36. [PMID: 14690521 DOI: 10.1046/j.1471-4159.2003.02143.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Proteins which recognize the two messengers phosphatidylinositol 3,4,5-trisphosphate (PtdInsP3), a membrane lipid, and inositol 1,3,4,5-tetrakisphosphate (InsP4), a water-soluble ligand, play important roles by integrating external stimuli, which lead to differentiation, cell death or survival. p42IP4, a PtdInsP3/InsP4-binding protein, is predominantly expressed in brain. The recently described centaurin alpha2 of similar molecular mass which is 58% identical and 75% homologous to the human p42IP4 orthologue, is expressed rather ubiquitously in many tissues. Here, elucidating the gene structure for both proteins, we found the human gene for centaurin alpha2 located on chromosome 17, position 17q11.2, near to the NF1 locus, and human p42IP4 on chromosome 7, position 7p22.3. The two isoforms, which both have 11 exons and conserved exon/intron transitions, seem to result from gene duplication. Furthermore, we studied binding of the two second messengers, PtdInsP3 and InsP4, and subcellular localization of the two proteins. Using recombinant baculovirus we expressed centaurin alpha2 and p42IP4 in Sf9 cells and purified the proteins to homogeneity. Recombinant centaurin alpha2 bound both InsP4 and PtdInsP3 equally well in vitro. Furthermore, fusion proteins of centaurin alpha2 and p42IP4, respectively, with the green fluorescent protein (GFP) were expressed in HEK 293 cells to visualize subcellular distribution. In contrast to p42IP4, which was distributed throughout the cell, centaurin alpha2 was concentrated at the plasma membrane already in unstimulated cells. The protein centaurin alpha2 was released from the membrane upon addition of wortmannin, which inhibits PI3-kinase. p42IP4, however, translocated to plasma membrane upon growth factor stimulation. Thus, in spite of the high homology between centaurin alpha2 and p42IP4 and comparable affinities for InsP4 and PtdInsP3, both proteins showed clear differences in subcellular distribution. We suggest a model, which is based on the difference in phosphoinositide binding stoichiometry of the two proteins, to account for the difference in subcellular localization.
Collapse
Affiliation(s)
- Theodor Hanck
- Institut für Neurobiochemie, Medizinische Fakultät, Otto-von-Guericke Universität Magdeburg, Magdeburg, Germany
| | | | | | | |
Collapse
|
15
|
Venkateswarlu K, Brandom KG, Lawrence JL. Centaurin-alpha1 is an in vivo phosphatidylinositol 3,4,5-trisphosphate-dependent GTPase-activating protein for ARF6 that is involved in actin cytoskeleton organization. J Biol Chem 2003; 279:6205-8. [PMID: 14625293 DOI: 10.1074/jbc.c300482200] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The ADP-ribosylation factor (ARF) 6 small GTPase regulates vesicle trafficking and cytoskeletal actin reorganization. The GTPase-activating proteins (GAPs) catalyze the formation of inactive ARF6GDP. Centaurin-alpha1 contains an ARF GAP and two pleckstrin homology (PH) domains, which bind the second messenger phosphatidylinositol 3,4,5-trisphosphate (PIP3). Here, we show that centaurin-alpha1 specifically inhibits in vivo GTP loading of ARF6 and redistribution of ARF6 from the endosomal compartment to the plasma membrane, which are indicative of its activation. Centaurin-alpha1 also inhibited cortical actin formation in a PIP3-dependent manner. Moreover, the constitutively active mutant of ARF6, but not that of ARF1, reverses the inhibition of cortical actin formation by centaurin-alpha1. An artificially plasma membrane-targeted centaurin-alpha1 bypasses the requirement of PIP3 for its involvement in ARF6 inactivation, suggesting that PIP3 is required for recruitment of centaurin-alpha1 to the plasma membrane but not for its activity. Together, these data suggest that centaurin-alpha1 negatively regulates ARF6 activity by functioning as an in vivo PIP3-dependent ARF6 GAP.
Collapse
|
16
|
Venkateswarlu K. Interaction protein for cytohesin exchange factors 1 (IPCEF1) binds cytohesin 2 and modifies its activity. J Biol Chem 2003; 278:43460-9. [PMID: 12920129 DOI: 10.1074/jbc.m304078200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The ADP-ribosylation factor 6 (ARF6) small GTPase functions as a GDP/GTP-regulated switch in the pathways that stimulate actin reorganization and membrane ruffling. The formation of active ARF6GTP is stimulated by guanine nucleotide exchange factors (GEFs) such as cytohesins, which translocate to the plasma membrane in agonist-stimulated cells by binding the lipid second messenger phosphatidylinositol 3,4,5-trisphosphate through the pleckstrin homology domain with subsequent ARF6 activation. Using cytohesin 2 as bait in yeast two-hybrid screening, we have isolated a cDNA encoding a protein termed interaction protein for cytohesin exchange factors 1 (IPCEF1). Using yeast two-hybrid and glutathione S-transferase pull-down assays coupled with deletion mutational analysis, the specific domains required for the cytohesin 2-IPCEF1 interaction were mapped to the coiled-coil domain of cytohesin 2 and the C-terminal 121 amino acids of IPCEF1. IPCEF1 also interacts with the other members of the cytohesin family of ARF GEFs, suggesting that the interaction with IPCEF1 is highly conserved among the cytohesin family of ARF GEFs. The interaction of cytohesin 2 and IPCEF1 in mammalian cells was demonstrated by immunoprecipitation. Immunofluorescence analysis revealed that IPCEF1 co-localizes with cytohesin 2 to the cytosol in unstimulated cells and translocates to the plasma membrane via binding to cytohesin 2 in epidermal growth factor-stimulated cells. However, a deletion mutant of IPCEF1 that lacks the cytohesin 2 binding site failed to co-migrate with cytohesin 2 to the membrane in stimulated cells. The functional significance of the IPCEF1-cytohesin 2 interaction is demonstrated by showing that IPCEF1 increases the in vitro and in vivo stimulation of ARFGTP formation by cytohesin 2.
Collapse
|
17
|
Stricker R, Vandekerckhove J, Krishna MU, Falck JR, Hanck T, Reiser G. Oligomerization controls in tissue-specific manner ligand binding of native, affinity-purified p42IP4/centaurin α1 and cytohesins—proteins with high affinity for the messengers d-inositol 1,3,4,5-tetrakisphosphate/phosphatidylinositol 3,4,5-trisphosphate. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2003; 1651:102-15. [PMID: 14499594 DOI: 10.1016/s1570-9639(03)00241-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Several distinct receptor proteins for the second messengers Ins(1,3,4,5)P(4) and PtdIns(3,4,5)P(3) are already known, such as the brain-specific p42(IP4), which we have previously cloned from different species, and cytohesins. However, it is still unclear whether proteins interacting with phosphoinositide and inositolpolyphosphate second messengers are regulated differently in different tissues. Here, we investigated these native proteins for comparison also from rat lung cytosol and purified them by PtdIns(3,4,5)P(3) affinity chromatography. Proteins selectively binding Ins(1,3,4,5)P(4) with high affinity also showed high affinity and specificity towards PtdIns(3,4,5)P(3). In lung cytosol, two prominent protein bands were found in the eluate from a PtdIns(3,4,5)P(3) affinity column. We identified these proteins by mass spectrometry as the cytohesin family of Arf guanosine nucleotide exchange factors (cytohesin 1, ARNO, GRP-1) and as Bruton's tyrosine kinase. Western blot analysis indicated that p42(IP4) was present in lung only at very low concentrations. Applying the affinity purification scheme established for rat lung cytosol to cytosol from rat brain, however, yielded only p42(IP4). We identified cytohesins in rat brain by Western blotting and PCR, but cytohesins surprisingly did not bind to the PtdIns(3,4,5)P(3)-affinity column. Gel filtration experiments of brain cytosol revealed that brain cytohesins are bound to large molecular weight complexes (150 to more than 500 kDa). Thus, we hypothesize that this finding explains why brain cytohesins apparently do not bind the inositolphosphate ligand. In lung cytosol, on the other hand, cytohesins occur as dimers. Gel filtration also showed that p42(IP4) in brain cytosol occurs as a monomer. Thus, oligomerization (homomeric or heteromeric) of InsP(4)/PtdInsP(3) binding proteins can modulate their function in a tissue-dependent manner because it can modify their ability to interact with the ligands.
Collapse
Affiliation(s)
- Rolf Stricker
- Institut für Neurobiochemie, Medizinische Fakultät der Otto-von-Guericke-Universität Magdeburg, 39120 Magdeburg, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Zemlickova E, Dubois T, Kerai P, Clokie S, Cronshaw AD, Wakefield RID, Johannes FJ, Aitken A. Centaurin-alpha(1) associates with and is phosphorylated by isoforms of protein kinase C. Biochem Biophys Res Commun 2003; 307:459-65. [PMID: 12893243 DOI: 10.1016/s0006-291x(03)01187-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Centaurin-alpha(1) is a member of the family of ADP-ribosylation factors (ARF) GTPase activating proteins (GAPs), although ARF GAP activity has not yet been demonstrated. The human homologue, centaurin-alpha(1) functionally complements the ARF GAP activity of Gcs1 in yeast. Although Gcs1 is involved in the formation of actin filaments in vivo, the function of centaurin remains elusive. We have identified a number of novel centaurin-alpha(1) binding partners; including CKIalpha and nucleolin. In this report, we have focused on the interaction of centaurin-alpha(1) with PKC. All groups of PKC associate directly through their cysteine rich domains. Centaurin-alpha(1) is also a substrate for all PKC classes and we have identified the two sites of phosphorylation. This is the first report of a kinase that phosphorylates centaurin-alpha(1).
Collapse
Affiliation(s)
- Eva Zemlickova
- University of Edinburgh, School of Biomedical and Clinical Laboratory Sciences, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Aggensteiner M, Reiser G. Expression of the brain-specific membrane adapter protein p42IP4/centaurin alpha, a Ins(1,3,4,5)P4/PtdIns(3,4,5)P3 binding protein, in developing rat brain. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2003; 142:77-87. [PMID: 12694946 DOI: 10.1016/s0165-3806(03)00033-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Inositolphosphates and phosphatidylinositides are important second messengers. Previously p42(IP4), a protein with high affinity for both Ins(1,3,4,5)P(4) and PtdIns(3,4,5)P(3) has been characterized in our laboratory. In the present study mRNA levels of p42(IP4) were quantified during development (ages: 7, 14, 21 days and adult) by means of ribonuclease protection assay in various rat brain regions (cerebellum, cortex, striatum, thalamus, hypothalamus, olfactory bulb, hippocampus and tectum (superior and inferior colliculus)). A high level of p42(IP4) mRNA was detected in the cortex (ca. 1 pg specific RNA per microg of total RNA) which stayed highly independent of the age of the animals. In hippocampus and in the thalamus, p42(IP4) mRNA levels were comparable to those in the cortex in the first and second week postnatally, but decreased to lower levels in the adult brain. In striatum, the mRNA increased, albeit less intensely than in hippocampus and thalamus, until day 21 postnatally, and then decreased in the adult rat brain. Cerebellar p42(IP4) mRNA showed a slow increase within the first 3 weeks postnatally, and remained rather high in the adult brain. The protein expression of p42(IP4), tested within the same samples by Western blot staining, was consistent with mRNA values. For comparison, glutamic acid decarboxylase (isoforms GAD65/GAD67), an enzyme, for which some regional brain specific distribution is already known, was also examined. The mRNA levels of GAD and its developmental regulation clearly differed from that of p42(IP4). In summary, p42(IP4) expressed in several neuronal cell types, did not seem to be restricted to specific developmental stages, but the high absolute expression levels at all developmental stages indicated that p42(IP4) is a protein fundamental for neuronal functioning.
Collapse
Affiliation(s)
- Michael Aggensteiner
- Institut für Neurobiochemie, Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | | |
Collapse
|
20
|
Dubois T, Zemlickova E, Howell S, Aitken A. Centaurin-alpha 1 associates in vitro and in vivo with nucleolin. Biochem Biophys Res Commun 2003; 301:502-8. [PMID: 12565890 DOI: 10.1016/s0006-291x(02)03010-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Centaurin-alpha(1) was originally described as a binding partner for phosphoinositides. In spite of the presence of a putative ADP-ribosylation factor (ARF) GTPase-activating protein (GAP) domain, no ARF-GAP activity has been attributed to centaurin-alpha(1) so far. Thus the function of this protein remains to be determined. In order to better understand its intracellular role, we aimed to identify centaurin-alpha(1) partners. Using affinity chromatography followed by mass spectrometry analysis, we identified several potential centaurin-alpha(1) protein partners. Nucleolin, a nucleolar protein involved in ribosome biosynthesis, was the main centaurin-alpha(1) interacting protein. The interaction between centaurin-alpha(1) and nucleolin was confirmed by Western blot analysis and GST pull down assays. Moreover, we have shown that ectopically expressed centaurin-alpha(1) associates in vivo with endogenous nucleolin in human embryonic kidney 293 cells. In addition, the association between nucleolin and centaurin-alpha(1) was disrupted by RNAse treatment, indicating that RNA integrity was necessary for their binding. This suggested that centaurin-alpha(1) was part of a ribonucleoprotein complex.
Collapse
Affiliation(s)
- Thierry Dubois
- Division of Biomedical and Clinical Laboratory Sciences, The University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK.
| | | | | | | |
Collapse
|
21
|
Reiser G, Bernstein HG. Neurons and plaques of Alzheimer's disease patients highly express the neuronal membrane docking protein p42IP4/centaurin alpha. Neuroreport 2002; 13:2417-9. [PMID: 12499840 DOI: 10.1097/00001756-200212200-00008] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The protein p42(IP4) (also called centaurin alpha), identified as a brain-specific InsP4/PtdInsP3 (PIP3)-binding protein, has been shown to be localized in human brain, specifically expressed in neurons. Several casein kinases have been found to be involved in Alzheimer's disease (AD) pathology. Since casein kinase I was reported to possess a binding domain for p42(IP4), we here investigated the expression and localization of p42(IP4) in AD brains. In cortical neurons of AD brains intracellular immunostaining for p42(IP4) exceeded the level seen in these neurons of normal brain. Statistically, significantly more p42(IP4)-immunoreactive neurons were found in temporal and angular cortex of AD patients as compared to control brain. Mostly impressively, neuritic plaques displayed a very prominent signal. Thus, we suggest that the up-regulated p42(IP4) in AD neurons may serve as a docking protein to recruit signaling molecules such as different subtypes of casein kinase I to the plasma membrane. This is the first indication for a functional interaction of these protein in possible neuronal damage. Therefore proteins such as p42(IP4), central players in signaling, may be appropriate targets for preventing neurodegenerative processes.
Collapse
Affiliation(s)
- Georg Reiser
- Klinik für Psychiatrie, Psychotherapie und Psychosomatische Medizin, Medizinische Fakultät, Leipziger Strasse 44, 39120 Magdeburg, Germany
| | | |
Collapse
|
22
|
Neri LM, Borgatti P, Capitani S, Martelli AM. The nuclear phosphoinositide 3-kinase/AKT pathway: a new second messenger system. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1584:73-80. [PMID: 12385889 DOI: 10.1016/s1388-1981(02)00300-1] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Lipid second messengers, particularly those derived from the polyphosphoinositide cycle, play a pivotal role in several cell signaling networks. Phosphoinositide 3-kinases (PI3Ks) generate specific inositol lipids that have been implicated in a plethora of cell functions. One of the best-characterized targets of PI3K lipid products is the serine/threonine protein kinase Akt. Recent findings have implicated Akt in cancer progression because it stimulates cell proliferation and suppresses apoptosis. Evidence accumulated over the past 15 years has highlighted the presence of an autonomous nuclear inositol lipid metabolism, and suggests that lipid molecules are important components of signaling pathways operating within the nucleus. PI3Ks, their lipid products, and Akt have also been identified at the nuclear level. In this review, we shall summarize the most updated findings about these molecules in relationship with the nuclear compartment and provide an overview of the possible mechanisms by which they regulate important cell functions.
Collapse
Affiliation(s)
- Luca M Neri
- Dipartimento di Morfologia ed Embriologia, Sezione di Anatomia Umana, Università di Ferrara, via Fossato di Mortara 66, 44100 Ferrara, Italy.
| | | | | | | |
Collapse
|
23
|
Sly BJ, Hazel JC, Popodi EM, Raff RA. Patterns of gene expression in the developing adult sea urchin central nervous system reveal multiple domains and deep-seated neural pentamery. Evol Dev 2002; 4:189-204. [PMID: 12054292 DOI: 10.1046/j.1525-142x.2002.02002.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The adult sea urchin central nervous system (CNS) is composed of five radial nerve cords connected to a circular nerve ring. Although much is known about the molecular mechanisms underlying the development and function of the nervous systems of many invertebrate and vertebrate species, virtually nothing is known about these processes in echinoderms. We have isolated a set of clones from a size-selected cDNA library prepared from the nervous system of the sea urchin Heliocidaris erythrogramma for use as probes. A total of 117 expressed sequence clones were used to search the GenBank database. Identified messages include genes that encode signaling proteins, cytoskeletal elements, cell surface proteins and receptors, cell proliferation and differentiation factors, transport and channel proteins, and a RNA DEAD box helicase. Expression was analyzed by RNA gel blot hybridization to document expression through development. Many of the genes have apparently neural limited expression and function, but some have been co-opted into new roles, notably associated with exocytotic events at fertilization. Localization of gene expression by whole-mount in situ hybridization shows that the morphologically simple sea urchin radial CNS exhibits complex organization into localized transcriptional domains. The transcription patterns reflect the morphological pentamery of the echinoderm CNS and provide no indication of an underlying functional bilateral symmetry in the CNS.
Collapse
Affiliation(s)
- Belinda J Sly
- Indiana Molecular Biology Institute and Department of Biology, Indiana University, Bloomington 47405, USA
| | | | | | | |
Collapse
|
24
|
Whitley P, Gibbard AM, Koumanov F, Oldfield S, Kilgour EE, Prestwich GD, Holman GD. Identification of centaurin-alpha2: a phosphatidylinositide-binding protein present in fat, heart and skeletal muscle. Eur J Cell Biol 2002; 81:222-30. [PMID: 12018390 DOI: 10.1078/0171-9335-00242] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We describe here the cloning, expression and characterisation of centaurin-alpha2 from a rat adipocyte cDNA library. The centaurin-alpha2 cDNA contains an open reading frame, which codes for a protein of 376 amino acids with predicted mass of 43.5 kDa. Centaurin-alpha2 shares 51-59% identity with centaurin-alpha1 proteins and has the same domain organisation, consisting of a predicted N-terminal ArfGAP domain followed by two successive pleckstrin homology domains. Despite the sequence similarity, there are a number of notable differences between the previously characterised centaurin-alpha1 proteins and the newly described centaurin-alpha2: (i) in vitro lipid binding experiments with centaurin-alpha2 do not reveal the same selectivity for phosphatidylinositol 3,4,5-trisphosphate over phosphatidylinositol 4,5-bisphosphate that has been shown for centaurin-alpha; (ii) unlike centaurin-alpha1 which is expressed mainly in the brain, centaurin-alpha2 has a broad tissue distribution, being particularly abundant in fat, heart and skeletal muscle; (iii) in contrast to centaurin-alpha1 which is found in both membrane and cytosolic fractions, endogenous centaurin-alpha2 is exclusively present in the dense membrane fractions of cell extracts, suggesting a constitutive membrane association. Insulin stimulation, which stimulates phosphatidylinositol 3,4,5-trisphosphate production, does not alter the subcellular distribution of centaurin-alpha2 between adipocyte membrane fractions. This observation is consistent with the lack of specificity of centaurin-alpha2 for phosphatidylinositol 3,4,5-trisphosphate over phosphatidylinositol 4,5-bisphosphate.
Collapse
Affiliation(s)
- Paul Whitley
- Department of Biology and Biochemistry, University of Bath, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
25
|
Sedehizade F, Hanck T, Stricker R, Horstmayer A, Bernstein HG, Reiser G. Cellular expression and subcellular localization of the human Ins(1,3,4,5)P(4)-binding protein, p42(IP4), in human brain and in neuronal cells. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 99:1-11. [PMID: 11869802 DOI: 10.1016/s0169-328x(01)00335-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In this study we describe for the human inositol-(1,3,4,5)-tetrakisphosphate (InsP4)-binding protein, p42IP4, the cellular distribution and subcellular localization in human brain and in transfected neuronal cells. The cDNA sequence of the human p42IP4 containing a single open reading frame yields a peptide of 374 amino acids with a calculated molecular mass of 43.4 kDa with a zinc-finger motif at the N-terminus, followed by two pleckstrin homology (PH) domains. Using a peptide-specific antiserum, p42IP4 protein was localized in a majority of neuronal cells of human brain sections. In the hypothalamus a subpopulation of paraventricular and infundibular nucleus neurons were strongly immunoreactive for p42IP4. In cortical areas the protein was predominantly found in large pyramidal cells. Some immunoreactivity for p42IP4 was also observed in the pyramidal cells of the hippocampal formation. Functional expression of p42IP4 protein in neuronal (NG108-15) and non-neuronal (CHO-K1) cells stably transfected with GFP-p42IP4 was shown in all cell fractions (homogenate, cytosol and membranes) by specific [3H]Ins(1,3,4,5)P4 binding activity, which correlated with p42IP4 protein detection by Western blot analysis. Using confocal laser scanning microscopy we showed that in NG108-15 and CHO-K1 cells stably transfected with GFP-p42IP4 the full length p42IP4 protein was localized in the cytoplasm, at the plasma membrane and in the nucleus. A deletion mutant of p42IP4 lacking the zinc finger domain resulted in solely a cytosolic and membrane localization but was not found in the nucleus. Thus we can conclude that human p42IP4 shows a region-specific localization in the human brain and the zinc finger motif within the protein is responsible for the localization of the protein in the cell nucleus.
Collapse
Affiliation(s)
- Fariba Sedehizade
- Institut für Neurobiochemie, Otto-von-Guericke Universität Magdeburg, Leipziger Strasse 44, 39120 Magdeburg, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Dubois T, Kerai P, Zemlickova E, Howell S, Jackson TR, Venkateswarlu K, Cullen PJ, Theibert AB, Larose L, Roach PJ, Aitken A. Casein kinase I associates with members of the centaurin-alpha family of phosphatidylinositol 3,4,5-trisphosphate-binding proteins. J Biol Chem 2001; 276:18757-64. [PMID: 11278595 DOI: 10.1074/jbc.m010005200] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mammalian casein kinases I (CKI) belong to a family of serine/threonine protein kinases involved in diverse cellular processes including cell cycle progression, membrane trafficking, circadian rhythms, and Wnt signaling. Here we show that CKIalpha co-purifies with centaurin-alpha(1) in brain and that they interact in vitro and form a complex in cells. In addition, we show that the association is direct and occurs through the kinase domain of CKI within a loop comprising residues 217-233. These residues are well conserved in all members of the CKI family, and we show that centaurin-alpha(1) associates in vitro with all mammalian CKI isoforms. To date, CKIalpha represents the first protein partner identified for centaurin-alpha(1). However, our data suggest that centaurin-alpha(1) is not a substrate for CKIalpha and has no effect on CKIalpha activity. Centaurin-alpha(1) has been identified as a phosphatidylinositol 3,4,5-trisphosphate-binding protein. Centaurin-alpha(1) contains a cysteine-rich domain that is shared by members of a newly identified family of ADP-ribosylation factor guanosine trisphosphatase-activating proteins. These proteins are involved in membrane trafficking and actin cytoskeleton rearrangement, thus supporting a role for CKIalpha in these biological events.
Collapse
Affiliation(s)
- T Dubois
- University of Edinburgh, Division of Biomedical and Clinical Laboratory Sciences, Hugh Robson Building, George Square, Edinburgh EH8 9XD
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Cocco L, Martelli AM, Gilmour RS, Rhee SG, Manzoli FA. Nuclear phospholipase C and signaling. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1530:1-14. [PMID: 11341954 DOI: 10.1016/s1388-1981(00)00169-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- L Cocco
- Cellular Signaling Laboratory, Department of Anatomical Sciences and Skeletal Muscle Pathophysiology, University of Bologna, Italy.
| | | | | | | | | |
Collapse
|
28
|
Lafferty AR, Torpy DJ, Stowasser M, Taymans SE, Lin JP, Huggard P, Gordon RD, Stratakis CA. A novel genetic locus for low renin hypertension: familial hyperaldosteronism type II maps to chromosome 7 (7p22). J Med Genet 2000; 37:831-5. [PMID: 11073536 PMCID: PMC1734468 DOI: 10.1136/jmg.37.11.831] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Familial hyperaldosteronism type II (FH-II) is caused by adrenocortical hyperplasia or aldosteronoma or both and is frequently transmitted in an autosomal dominant fashion. Unlike FH type I (FH-I), which results from fusion of the CYP11B1 and CYP11B2 genes, hyperaldosteronism in FH-II is not glucocorticoid remediable. A large family with FH-II was used for a genome wide search and its members were evaluated by measuring the aldosterone:renin ratio. In those with an increased ratio, FH-II was confirmed by fludrocortisone suppression testing. After excluding most of the genome, genetic linkage was identified with a maximum two point lod score of 3.26 at theta=0, between FH-II in this family and the polymorphic markers D7S511, D7S517, and GATA24F03 on chromosome 7, a region that corresponds to cytogenetic band 7p22. This is the first identified locus for FH-II; its molecular elucidation may provide further insight into the aetiology of primary aldosteronism.
Collapse
Affiliation(s)
- A R Lafferty
- Unit on Genetics and Endocrinology, Developmental Endocrinology Branch, National Institute of Child Health and Human Development, Building 10, Room 10N262, 10 Center Drive, MSC1862, Bethesda, Maryland 20892-1862, USA
| | | | | | | | | | | | | | | |
Collapse
|