1
|
Boldyreva LV, Evtushenko AA, Lvova MN, Morozova KN, Kiseleva EV. Underneath the Gut-Brain Axis in IBD-Evidence of the Non-Obvious. Int J Mol Sci 2024; 25:12125. [PMID: 39596193 PMCID: PMC11594934 DOI: 10.3390/ijms252212125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
The gut-brain axis (GBA) plays a pivotal role in human health and wellness by orchestrating complex bidirectional regulation and influencing numerous critical processes within the body. Over the past decade, research has increasingly focused on the GBA in the context of inflammatory bowel disease (IBD). Beyond its well-documented effects on the GBA-enteric nervous system and vagus nerve dysregulation, and gut microbiota misbalance-IBD also leads to impairments in the metabolic and cellular functions: metabolic dysregulation, mitochondrial dysfunction, cationic transport, and cytoskeleton dysregulation. These systemic effects are currently underexplored in relation to the GBA; however, they are crucial for the nervous system cells' functioning. This review summarizes the studies on the particular mechanisms of metabolic dysregulation, mitochondrial dysfunction, cationic transport, and cytoskeleton impairments in IBD. Understanding the involvement of these processes in the GBA may help find new therapeutic targets and develop systemic approaches to improve the quality of life in IBD patients.
Collapse
Affiliation(s)
- Lidiya V. Boldyreva
- Scientific-Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia;
| | - Anna A. Evtushenko
- Scientific-Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia;
| | - Maria N. Lvova
- Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.N.L.); (K.N.M.); (E.V.K.)
| | - Ksenia N. Morozova
- Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.N.L.); (K.N.M.); (E.V.K.)
| | - Elena V. Kiseleva
- Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.N.L.); (K.N.M.); (E.V.K.)
| |
Collapse
|
2
|
Salloum G, Bresnick AR, Backer JM. Macropinocytosis: mechanisms and regulation. Biochem J 2023; 480:335-362. [PMID: 36920093 DOI: 10.1042/bcj20210584] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/16/2023]
Abstract
Macropinocytosis is defined as an actin-dependent but coat- and dynamin-independent endocytic uptake process, which generates large intracellular vesicles (macropinosomes) containing a non-selective sampling of extracellular fluid. Macropinocytosis provides an important mechanism of immune surveillance by dendritic cells and macrophages, but also serves as an essential nutrient uptake pathway for unicellular organisms and tumor cells. This review examines the cell biological mechanisms that drive macropinocytosis, as well as the complex signaling pathways - GTPases, lipid and protein kinases and phosphatases, and actin regulatory proteins - that regulate macropinosome formation, internalization, and disposition.
Collapse
Affiliation(s)
- Gilbert Salloum
- Department of Molecular Pharamacology, Albert Einstein College of Medicine, Bronx, NY, U.S.A
| | - Anne R Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, U.S.A
| | - Jonathan M Backer
- Department of Molecular Pharamacology, Albert Einstein College of Medicine, Bronx, NY, U.S.A
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, U.S.A
| |
Collapse
|
3
|
McDermott MI, Wang Y, Wakelam MJO, Bankaitis VA. Mammalian phospholipase D: Function, and therapeutics. Prog Lipid Res 2019; 78:101018. [PMID: 31830503 DOI: 10.1016/j.plipres.2019.101018] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/08/2019] [Accepted: 10/14/2019] [Indexed: 01/23/2023]
Abstract
Despite being discovered over 60 years ago, the precise role of phospholipase D (PLD) is still being elucidated. PLD enzymes catalyze the hydrolysis of the phosphodiester bond of glycerophospholipids producing phosphatidic acid and the free headgroup. PLD family members are found in organisms ranging from viruses, and bacteria to plants, and mammals. They display a range of substrate specificities, are regulated by a diverse range of molecules, and have been implicated in a broad range of cellular processes including receptor signaling, cytoskeletal regulation and membrane trafficking. Recent technological advances including: the development of PLD knockout mice, isoform-specific antibodies, and specific inhibitors are finally permitting a thorough analysis of the in vivo role of mammalian PLDs. These studies are facilitating increased recognition of PLD's role in disease states including cancers and Alzheimer's disease, offering potential as a target for therapeutic intervention.
Collapse
Affiliation(s)
- M I McDermott
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, United States of America.
| | - Y Wang
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, United States of America; Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843-2128, United States of America
| | - M J O Wakelam
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - V A Bankaitis
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, United States of America; Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843-2128, United States of America; Department of Chemistry, Texas A&M University, College Station, Texas 77840, United States of America
| |
Collapse
|
4
|
Bruntz RC, Lindsley CW, Brown HA. Phospholipase D signaling pathways and phosphatidic acid as therapeutic targets in cancer. Pharmacol Rev 2015; 66:1033-79. [PMID: 25244928 DOI: 10.1124/pr.114.009217] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Phospholipase D is a ubiquitous class of enzymes that generates phosphatidic acid as an intracellular signaling species. The phospholipase D superfamily plays a central role in a variety of functions in prokaryotes, viruses, yeast, fungi, plants, and eukaryotic species. In mammalian cells, the pathways modulating catalytic activity involve a variety of cellular signaling components, including G protein-coupled receptors, receptor tyrosine kinases, polyphosphatidylinositol lipids, Ras/Rho/ADP-ribosylation factor GTPases, and conventional isoforms of protein kinase C, among others. Recent findings have shown that phosphatidic acid generated by phospholipase D plays roles in numerous essential cellular functions, such as vesicular trafficking, exocytosis, autophagy, regulation of cellular metabolism, and tumorigenesis. Many of these cellular events are modulated by the actions of phosphatidic acid, and identification of two targets (mammalian target of rapamycin and Akt kinase) has especially highlighted a role for phospholipase D in the regulation of cellular metabolism. Phospholipase D is a regulator of intercellular signaling and metabolic pathways, particularly in cells that are under stress conditions. This review provides a comprehensive overview of the regulation of phospholipase D activity and its modulation of cellular signaling pathways and functions.
Collapse
Affiliation(s)
- Ronald C Bruntz
- Department of Pharmacology (R.C.B., C.W.L., H.A.B.) and Vanderbilt Center for Neuroscience Drug Discovery (C.W.L.), Vanderbilt University Medical Center; Department of Chemistry, Vanderbilt Institute of Chemical Biology (C.W.L., H.A.B.); Vanderbilt Specialized Chemistry for Accelerated Probe Development (C.W.L.); and Department of Biochemistry, Vanderbilt-Ingram Cancer Center (H.A.B.), Vanderbilt University, Nashville, Tennessee
| | - Craig W Lindsley
- Department of Pharmacology (R.C.B., C.W.L., H.A.B.) and Vanderbilt Center for Neuroscience Drug Discovery (C.W.L.), Vanderbilt University Medical Center; Department of Chemistry, Vanderbilt Institute of Chemical Biology (C.W.L., H.A.B.); Vanderbilt Specialized Chemistry for Accelerated Probe Development (C.W.L.); and Department of Biochemistry, Vanderbilt-Ingram Cancer Center (H.A.B.), Vanderbilt University, Nashville, Tennessee
| | - H Alex Brown
- Department of Pharmacology (R.C.B., C.W.L., H.A.B.) and Vanderbilt Center for Neuroscience Drug Discovery (C.W.L.), Vanderbilt University Medical Center; Department of Chemistry, Vanderbilt Institute of Chemical Biology (C.W.L., H.A.B.); Vanderbilt Specialized Chemistry for Accelerated Probe Development (C.W.L.); and Department of Biochemistry, Vanderbilt-Ingram Cancer Center (H.A.B.), Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
5
|
Phosphatidylinositol 4,5-bisphosphate is a novel coactivator of the Pseudomonas aeruginosa cytotoxin ExoU. Infect Immun 2013; 81:2873-81. [PMID: 23716613 DOI: 10.1128/iai.00414-13] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ExoU is a potent phospholipase A2 effector protein secreted by the type III secretion system of Pseudomonas aeruginosa. By cleaving plasma membrane phospholipids, it causes rapid lysis of eukaryotic cells. However, ExoU does not exhibit activity on its own but instead requires eukaryotic cell cofactors for activation. Ubiquitin and ubiquitinated proteins have been shown to activate ExoU, but previous work suggested that other cofactors are also involved. In this study, we demonstrate that phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] is another important coactivator of ExoU. PI(4,5)P2 works synergistically with ubiquitin to greatly enhance the phospholipase A2 activity of ExoU. Distinct residues of ExoU were critical for activation by PI(4,5)P2 and by ubiquitin, indicating that these factors activate ExoU by discrete mechanisms. In support of the biological relevance of PI(4,5)P2 coactivation, a yeast mutant with reduced PI(4,5)P2 levels was less susceptible to the cytotoxic activity of ExoU. Together, these findings further elaborate the molecular mechanism of ExoU.
Collapse
|
6
|
Gomez-Cambronero J, Henkels KM. Cloning of PLD2 from baculovirus for studies in inflammatory responses. Methods Mol Biol 2012; 861:201-25. [PMID: 22426721 DOI: 10.1007/978-1-61779-600-5_13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The enzyme PLD hydrolyzes phosphodiester bonds of lipids in cell membranes. Phosphatidic acid, a chief product of PLD enzymatic activity, is a pleiotropic second messenger with key roles in membrane trafficking, cell invasion, cell growth, and anti-apoptosis. We describe in the present study molecular, cellular, and physiological methods to understand the mechanism of how the PLD2 isozyme regulates the process of inflammation. We describe here (1) a method that details phospholipase D2 (PLD2) cloning in the pBac expression vector, (2) the large-scale infection of Sf21 insect cells for protein production, (3) protein purification by TALON cobalt metal affinity matrix and subsequent assessment of PLD2 protein and lipase activity, (4) application of purified PLD2 protein for the study of Rac2 GTPase biology involving GTP binding by a pull-down assay and GTP/GDP exchange activity, (5) a method of PLD2 expression that involves mammalian cells, (6) a physiological application as relates to adhesion, chemotaxis, and phagocytosis, and (7) a model that integrates the results of a PLD-GTPase interaction from the molecular to the physiological contexts.
Collapse
Affiliation(s)
- Julian Gomez-Cambronero
- Department of Biochemistry and Molecular Biology, Wright State University School of Medicine, Dayton, OH, USA.
| | | |
Collapse
|
7
|
Gomez-Cambronero J. The exquisite regulation of PLD2 by a wealth of interacting proteins: S6K, Grb2, Sos, WASp and Rac2 (and a surprise discovery: PLD2 is a GEF). Cell Signal 2011; 23:1885-95. [PMID: 21740967 PMCID: PMC3204931 DOI: 10.1016/j.cellsig.2011.06.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 06/21/2011] [Indexed: 11/28/2022]
Abstract
Phospholipase D (PLD) catalyzes the conversion of the membrane phospholipid phosphatidylcholine to choline and phosphatidic acid (PA). PLD's mission in the cell is two-fold: phospholipid turnover with maintenance of the structural integrity of cellular/intracellular membranes and cell signaling through PA and its metabolites. Precisely, through its product of the reaction, PA, PLD has been implicated in a variety of physiological cellular functions, such as intracellular protein trafficking, cytoskeletal dynamics, chemotaxis of leukocytes and cell proliferation. The catalytic (HKD) and regulatory (PH and PX) domains were studied in detail in the PLD1 isoform, but PLD2 was traditionally studied in lesser detail and much less was known about its regulation. Our laboratory has been focusing on the study of PLD2 regulation in mammalian cells. Over the past few years, we have reported, in regards to the catalytic action of PLD, that PA is a chemoattractant agent that binds to and signals inside the cell through the ribosomal S6 kinases (S6K). Regarding the regulatory domains of PLD2, we have reported the discovery of the PLD2 interaction with Grb2 via Y169 in the PX domain, and further association to Sos, which results in an increase of de novo DNA synthesis and an interaction (also with Grb2) via the adjacent residue Y179, leading to the regulation of cell ruffling, chemotaxis and phagocytosis of leukocytes. We also present the complex regulation by tyrosine phosphorylation by epidermal growth factor receptor (EGF-R), Janus Kinase 3 (JAK3) and Src and the role of phosphatases. Recently, there is evidence supporting a new level of regulation of PLD2 at the PH domain, by the discovery of CRIB domains and a Rac2-PLD2 interaction that leads to a dual (positive and negative) effect on its enzymatic activity. Lastly, we review the surprising finding of PLD2 acting as a GEF. A phospholipase such as PLD that exists already in the cell membrane that acts directly on Rac allows a quick response of the cell without intermediary signaling molecules. This provides only the latest level of PLD2 regulation in a field that promises newer and exciting advances in the next few years.
Collapse
Affiliation(s)
- Julian Gomez-Cambronero
- Department of Biochemistry and Molecular Biology, Wright State University School of Medicine, Dayton, OH 45435, USA.
| |
Collapse
|
8
|
Jeon H, Kwak D, Noh J, Lee MN, Lee CS, Suh PG, Ryu SH. Phospholipase D2 induces stress fiber formation through mediating nucleotide exchange for RhoA. Cell Signal 2011; 23:1320-6. [PMID: 21440060 DOI: 10.1016/j.cellsig.2011.03.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 03/15/2011] [Indexed: 12/23/2022]
Abstract
Phospholipase D (PLD) is involved in diverse cellular processes including cell movement, adhesion, and vesicle trafficking through cytoskeletal rearrangements. However, the mechanism by which PLD induces cytoskeletal reorganization is still not fully understood. Here, we describe a new link to cytoskeletal changes that is mediated by PLD2 through direct nucleotide exchange on RhoA. We found that PLD2 induces RhoA activation independent of its lipase activity. PLD2 directly interacted with RhoA, and the PX domain of PLD2 specifically recognized nucleotide-free RhoA. Finally, we found that the PX domain of PLD2 has guanine nucleotide-exchange factor (GEF) activity for RhoA in vitro. In addition, we verified that overexpression of the PLD2-PX domain induces RhoA activation, thereby provoking stress fiber formation. Together, our findings suggest that PLD2 functions as an upstream regulator of RhoA, which enables us to understand how PLD2 regulates cytoskeletal reorganization in a lipase activity-independent manner.
Collapse
Affiliation(s)
- Hyeona Jeon
- Division of Molecular and Life Sciences, Pohang University of Science and Technology, Pohang, 790-784, South Korea
| | | | | | | | | | | | | |
Collapse
|
9
|
Protein kinase Cdelta-mediated phosphorylation of phospholipase D controls integrin-mediated cell spreading. Mol Cell Biol 2010; 30:5086-98. [PMID: 20733000 DOI: 10.1128/mcb.00443-10] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Integrin signaling plays critical roles in cell adhesion, spreading, and migration, and it is generally accepted that to regulate these integrin functions accurately, localized actin remodeling is required. However, the molecular mechanisms that control the targeting of actin regulation molecules to the proper sites are unknown. We previously demonstrated that integrin-mediated cell spreading and migration on fibronectin are dependent on the localized activation of phospholipase D (PLD). However, the mechanism underlying PLD activation by integrin is largely unknown. Here we demonstrate that protein kinase Cδ (PKCδ) is required for integrin-mediated PLD signaling. After integrin stimulation, PKCδ is activated and translocated to the edges of lamellipodia, where it colocalizes with PLD2. The abrogation of PKCδ activity inhibited integrin-induced PLD activation and cell spreading. Finally, we show that Thr566 of PLD2 is directly phosphorylated by PKCδ and that PLD2 mutation in this region prevents PLD2 activation, PLD2 translocation to the edge of lamellipodia, Rac translocation, and cell spreading after integrin activation. Together, these results suggest that PKCδ is a primary regulator of integrin-mediated PLD activation via the direct phosphorylation of PLD, which is essential for directing integrin-induced cell spreading.
Collapse
|
10
|
Mansfeld J, Ulbrich-Hofmann R. Modulation of phospholipase D activity in vitro. Biochim Biophys Acta Mol Cell Biol Lipids 2009; 1791:913-26. [DOI: 10.1016/j.bbalip.2009.03.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Revised: 03/03/2009] [Accepted: 03/04/2009] [Indexed: 11/30/2022]
|
11
|
Kanaho Y, Funakoshi Y, Hasegawa H. Phospholipase D signalling and its involvement in neurite outgrowth. Biochim Biophys Acta Mol Cell Biol Lipids 2009; 1791:898-904. [DOI: 10.1016/j.bbalip.2009.03.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Revised: 03/12/2009] [Accepted: 03/19/2009] [Indexed: 11/26/2022]
|
12
|
|
13
|
Exton JH. In Search of the Message. J Biol Chem 2008; 283:14901-9. [DOI: 10.1074/jbc.x800001200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
14
|
|
15
|
|
16
|
Gayral S, Déléris P, Laulagnier K, Laffargue M, Salles JP, Perret B, Record M, Breton-Douillon M. Selective activation of nuclear phospholipase D-1 by g protein-coupled receptor agonists in vascular smooth muscle cells. Circ Res 2006; 99:132-9. [PMID: 16778131 DOI: 10.1161/01.res.0000232323.86227.8b] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent studies highlight the existence of an autonomous nuclear lipid metabolism related to cellular proliferation. However, the importance of nuclear phosphatidylcholine (PC) metabolism is poorly understood. Therefore, we were interested in nuclear PCs as a source of second messengers and, particularly, nuclear phospholipase D (PLD) identification in membrane-free nuclei isolated from pig aorta vascular smooth muscle cells (VSMCs). Using immunoblot experiment, in vitro PLD assay with fluorescent substrate and confocal microscopy analysis, we demonstrated that only PLD1 is expressed in VSMC nucleus, whereas PLD1 and PLD2 are present in VSMC. Inhibition of RhoA and protein kinase Czeta (PKCzeta) by C3-exoenzyme and PKCzeta pseudosubstrate inhibitor, respectively, conducted a decrease of phosphatidylethanol production. On the other hand, treatment of intact VSMCs, but not nuclei, with phosphoinositide 3-kinase (PI3K) inhibitors prevented partially nuclear PLD1 activity, indicating for the first time that PI3K may have a role in nuclear PLD regulation. In addition, lysophosphatidic acid (LPA) or angiotensin II treatment of VSMCs resulted in an increase of intranuclear PLD activity, whereas platelet-derived growth factor and epidermal growth factor have no significant effect. Moreover, pertussis toxin induced a decrease of LPA-stimulated nuclear PLD1 activity, suggesting that heterotrimeric G(i)/G(0) protein involvement in intranuclear PLD1 regulation. We also show that LPA-induced nuclear PLD1 activation implied PI3K/PKCzeta pathway activation and PKCzeta nuclear translocation as well as nuclear RhoA activation. Thus, the characterization of an endogenous PLD1 that could regulate PC metabolism inside VSMC nucleus provides a new role for this enzyme in control of vascular fibroproliferative disorders.
Collapse
Affiliation(s)
- Stéphanie Gayral
- Département Lipoprotéines and Médiateurs Lipidiques, CPTP, INSERM Unité 563, Bâtiment C, CHU Purpan, BP 3028, 31024 Toulouse Cedex 3, France
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Henage LG, Exton JH, Brown HA. Kinetic analysis of a mammalian phospholipase D: allosteric modulation by monomeric GTPases, protein kinase C, and polyphosphoinositides. J Biol Chem 2006; 281:3408-17. [PMID: 16339153 PMCID: PMC3800466 DOI: 10.1074/jbc.m508800200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In mammalian cells, phospholipase D activity is tightly regulated by diverse cellular signals, including hormones, neurotransmitters, and growth factors. Multiple signaling pathways converge upon phospholipase D to modulate cellular actions, such as cell growth, shape, and secretion. We examined the kinetics of protein kinase C and G-protein regulation of mammalian phospholipase D1 (PLD1) in order to better understand interactions between PLD1 and its regulators. Activation by Arf-1, RhoA, Rac1, Cdc42, protein kinase Calpha, and phosphatidylinositol 4,5-bisphosphate displayed surface dilution kinetics, but these effectors modulated different kinetic parameters. PKCalpha activation of PLD1 involves N- and C-terminal PLD domains. Rho GTPases were binding activators, enhancing the catalytic efficiency of a purified PLD1 catalytic domain via effects on Km. Arf-1, a catalytic activator, stimulated PLD1 by enhancing the catalytic constant, kcat. A kinetic description of PLD1 activation by multiple modulators reveals a mechanism for apparent synergy between activators. Synergy was observed only when PLD1 was simultaneously stimulated by a binding activator and a catalytic activator. Surprisingly, synergistic activation was steeply dependent on phosphatidylinositol 4,5-bisphosphate and phosphatidylcholine. Together, these findings suggest a role for PLD1 as a signaling node, in which integration of convergent signals occurs within discrete locales of the cellular membrane.
Collapse
Affiliation(s)
- Lee G. Henage
- Department of Pharmacology, Institute for Chemical Biology, and Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232-6600
| | - John H. Exton
- Department of Pharmacology, Institute for Chemical Biology, and Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232-6600
| | - H. Alex Brown
- Department of Pharmacology, Institute for Chemical Biology, and Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232-6600
| |
Collapse
|
18
|
Kook S, Exton JH. Identification of interaction sites of protein kinase Cα on phospholipase D1. Cell Signal 2005; 17:1423-32. [PMID: 15951158 DOI: 10.1016/j.cellsig.2005.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Revised: 02/22/2005] [Accepted: 03/03/2005] [Indexed: 11/29/2022]
Abstract
Phospholipase D (PLD) is regulated by many factors, including protein kinase C (PKC) and small G-proteins of the Rho and ADP-ribosylation factor families. Previous studies revealed that the activation of PLD1 by phorbol ester is associated with the binding of PKCalpha to a site in the N-terminus of PLD1. The purpose of the present study was to determine this site more precisely. Immunoprecipitation with a series of four PLD1 deletion mutants confirmed that PKCalpha strongly interacted with the amino acid sequence 1-318 at the N-terminus of PLD1 and weakly with the sequence 841-1036 at the C-terminus. Further immunoprecipitation studies with deletion mutants of the 1-318 and 1-215 PLD1 fragments revealed that there were binding sites in the 1-49 N-terminal sequence and also in the 216-318 sequence containing the PH domain. Studies of N-terminal deletion mutants of full-length PLD1 confirmed the presence of a binding site in the 1-49 sequence and a further site in the 1-318 sequence. Both deletion mutants showed impaired activation by PKCalpha in vivo, but unchanged activation by active V(14)RhoA. These findings identify the 1-49 sequence is a major binding/activation site for PKCalpha on PLD1, but also indicate involvement of the PH domain.
Collapse
Affiliation(s)
- Seunghyi Kook
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | |
Collapse
|
19
|
Kuan YH, Lin RH, Tsao LT, Chen YL, Tzeng CC, Wang JP. Inhibition of phospholipase D activation by CYL-26z in formyl peptide-stimulated neutrophils involves the blockade of RhoA activation. Biochem Pharmacol 2005; 70:901-10. [PMID: 16024001 DOI: 10.1016/j.bcp.2005.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Revised: 06/02/2005] [Accepted: 06/08/2005] [Indexed: 01/21/2023]
Abstract
5-[4-Acridin-9-ylamino]phenyl]-5-methyl-3-methylenedihydrofuran-2-one (CYL-26z) inhibited the formyl-Met-Leu-Phe (fMLP)-stimulated phospholipase D (PLD) activity, which was assessed by the production of phosphatidylethanol (PEt) in the presence of ethanol, in rat neutrophils (IC50 1.2+/-0.2 microM). CYL-26z caused a slight but significant attenuation of the global protein tyrosine phosphorylation stimulated by fMLP only at concentrations of CYL-26z up to 30 microM. CYL-26z blocked the membrane recruitment of protein kinase C-alpha (PKC-alpha) at concentrations of CYL-26z > or =3 microM, but failed to affect the membrane association of PKC-betaI and -betaII. The translocation of RhoA to the membrane was attenuated by CYL-26z (IC50 3.8+/-0.8 microM) in fMLP-stimulated neutrophils, whereas CYL-26z caused no significant inhibition of the membrane recruitment of ADP-ribosylation factor (Arf). CYL-26z inhibited the activation of RhoA and dissociation of the RhoA-Rho guanine nucleotide dissociation inhibitor (GDI) complex in fMLP-stimulated neutrophils (IC50 1.8+/-1.0 microM and 1.8+/-0.9 microM, respectively). In a cell-free system, CYL-26z effectively attenuated the membrane association of RhoA in response to GTPgammaS (IC50 1.3+/-0.5 microM). In contrast, the GTPgammaS-stimulated translocation of Arf to membrane was suppressed only at concentrations of CYL-26z up to 30 microM. CYL-26z inhibited the fMLP-stimulated membrane expression of CD11b, CD45 and CD63, and the release of lysozyme and beta-glucuronidase. These results indicate that CYL-26z inhibited the fMLP-stimulated PLD activity, mainly through the blockade of RhoA activation, and degranulation in rat neutrophils.
Collapse
Affiliation(s)
- Yu-Hsiang Kuan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
20
|
Lee JS, Kim JH, Jang IH, Kim HS, Han JM, Kazlauskas A, Yagisawa H, Suh PG, Ryu SH. Phosphatidylinositol (3,4,5)-trisphosphate specifically interacts with the phox homology domain of phospholipase D1 and stimulates its activity. J Cell Sci 2005; 118:4405-13. [PMID: 16179605 DOI: 10.1242/jcs.02564] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Phospholipase D (PLD), which catalyzes the hydrolysis of phosphatidylcholine to phosphatidic acid and choline, plays key roles in cellular signal transduction by mediating extracellular stimuli including hormones, growth factors, neurotransmitters, cytokines and extracellular matrix molecules. The molecular mechanisms by which domains regulate the activity of PLD - especially the phox homology (PX) domain - have not been fully elucidated. In this study, we have examined the properties of the PX domains of PLD1 and PLD2 in terms of phosphoinositide binding and PLD activity regulation. Interestingly, the PX domain of PLD1, but not that of PLD2, was found to specifically interact with phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P3). We found that mutation of the conserved arginine at position 179 of the PLD1 PX domain to lysine or to alanine (R179A or R179K, respectively) disrupts PtdIns(3,4,5)P3 binding. In NIH-3T3 cells, the EGFP-PLD1 PX wild-type domain, but not the two mutants, localized to the plasma membrane after 5-minute treatment with platelet-derived growth factor (PDGF). The enzymatic activity of PLD1 was stimulated by adding PtdIns(3,4,5)P3 in vitro. Treatment with PDGF resulted in the significant increase of PLD1 activity and phosphorylation of the downstream extracellular signal-regulated kinases (ERKs), which was blocked by pre-treatment of HEK 293 cells with phosphoinositide 3-kinase (PI3K) inhibitor after the endogenous PLD2 had been depleted by siRNA specific for PLD2. Nevertheless, both PLD1 mutants (which cannot interact with PtdIns(3,4,5)P3) did not respond to treatment with PDGF. Moreover, PLD1 was activated in HepG2 cells stably expressing the Y40/51 mutant of PDGF receptor that is required for the binding with PI3K. Our results suggest that the PLD1 PX domain enables PLD1 to mediate signal transduction via ERK1/2 by providing a direct binding site for PtdIns(3,4,5)P3 and by activating PLD1.
Collapse
Affiliation(s)
- Jun Sung Lee
- Division of Molecular and Life Science, Pohang University of Science and Technology, Pohang 790-784, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Hiroyama M, Exton JH. Studies of the roles of ADP-ribosylation factors and phospholipase D in phorbol ester-induced membrane ruffling. J Cell Physiol 2005; 202:608-22. [PMID: 15389577 DOI: 10.1002/jcp.20156] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In this study, we have explored the roles of ADP-ribosylation factors (ARFs), phospholipase D (PLD) isozymes, and arfaptins in phorbol ester (PMA)-induced membrane ruffling in HeLa cells. PMA stimulation induced ruffling and translocated cortactin to the plasma membrane. The cortactin translocation was inhibited by dominant negative (DN)-ARF6, DN-ARF1, and DN-Rac1, but not by DN-RhoA and DN-Cdc42. The inability of DN-forms of ARF6, ARF1, and Rac1 to affect PLD activity in response to PMA indicated that this enzyme was not activated via these small G proteins and that its activation was not essential for the induction of ruffling. Endogenous-ARF1, -ARF6, and -Rac1 existed in the ruffling region along with cortactin after PMA stimulation. DN-ARF1 had no effect on the ruffling induced by DA-ARF6 or DA-Rac1, and DN-ARF6 had no effect on that induced by DA-ARF1 or DA-Rac1. On the other hand DN-Rac1 suppressed the effect of DA-ARF6 but not that of DA-ARF1. These results suggest that PMA causes membrane ruffling via an ARF6-Rac1 pathway and also an ARF1 pathway operating in parallel. Overexpression of PLD1 and PLD2 inhibited PMA-induced cortactin translocation and actin-cortactin complex formation, supporting the view that these enzymes are not required for ruffling, but actually suppress it. We conclude that PMA-induced membrane ruffling is caused via ARF6-Rac1 and ARF1 pathways operating in parallel and that PLD may be inhibitory.
Collapse
Affiliation(s)
- Masami Hiroyama
- Howard Hughes Medical Institute and Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
22
|
Chae YC, Lee S, Lee HY, Heo K, Kim JH, Kim JH, Suh PG, Ryu SH. Inhibition of Muscarinic Receptor-linked Phospholipase D Activation by Association with Tubulin. J Biol Chem 2005; 280:3723-30. [PMID: 15548524 DOI: 10.1074/jbc.m406987200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mammalian phospholipase D (PLD) is considered a key enzyme in the transmission signals from various receptors including muscarinic receptors. PLD activation is a rapid and transient process, but a negative regulator has not been found that inhibits signal-dependent PLD activation. Here, for the first time, we report that tubulin binding to PLD2 is an inhibition mechanism for muscarinic receptor-linked PLD2 activation. Tubulin was identified in an immunoprecipitated PLD2 complex from COS-7 cells by peptide mass fingerprinting. The direct interaction between PLD2 and tubulin was found to be mediated by a specific region of PLD2 (amino acids 476-612). PLD2 was potently inhibited (IC50 <10 nM) by tubulin binding in vitro. In cells, the interaction between PLD2 and tubulin was increased by the microtubule disrupting agent nocodazole and reduced by the microtubule stabilizing agent Taxol. Moreover, PLD2 activity was found to be inversely correlated with the level of monomeric tubulin. In addition, we found that interaction with and the inhibition of PLD2 by monomeric tubulin is important for the muscarinic receptor-linked PLD signaling pathway. Interaction between PLD2 and tubulin was increased only after 1-2 min of carbachol stimulation when carbachol-stimulated PLD2 activity was decreased. The expression of the tubulin binding region of PLD2 blocked the later decrease in carbachol-induced PLD activity by masking tubulin binding. Taken together, these results indicate that an increase in local membrane monomeric tubulin concentration inhibits PLD2 activity, and provides a novel mechanism for the inhibition of muscarinic receptor-induced PLD2 activation by interaction with tubulin.
Collapse
Affiliation(s)
- Young Chan Chae
- Division of Molecular and Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Stahelin RV, Ananthanarayanan B, Blatner NR, Singh S, Bruzik KS, Murray D, Cho W. Mechanism of Membrane Binding of the Phospholipase D1 PX Domain. J Biol Chem 2004; 279:54918-26. [PMID: 15475361 DOI: 10.1074/jbc.m407798200] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mammalian phospholipases D (PLD), which catalyze the hydrolysis of phosphatidylcholine to phosphatidic acid (PA), have been implicated in various cell signaling and vesicle trafficking processes. Mammalian PLD1 contains two different membrane-targeting domains, pleckstrin homology and Phox homology (PX) domains, but the precise roles of these domains in the membrane binding and activation of PLD1 are still unclear. To elucidate the role of the PX domain in PLD1 activation, we constructed a structural model of the PX domain by homology modeling and measured the membrane binding of this domain and selected mutants by surface plasmon resonance analysis. The PLD1 PX domain was found to have high phosphoinositide specificity, i.e. phosphatidylinositol 3,4,5-trisphosphate (PtdIns-(3,4,5)P(3)) >> phosphatidylinositol 3-phosphate > phosphatidylinositol 5-phosphate >> other phosphoinositides. The PtdIns(3,4,5)P(3) binding was facilitated by the cationic residues (Lys(119), Lys(121), and Arg(179)) in the putative binding pocket. Consistent with the model structure that suggests the presence of a second lipid-binding pocket, vesicle binding studies indicated that the PLD1 PX domain could also bind with moderate affinity to PA, phosphatidylserine, and other anionic lipids, which were mediated by a cluster of cationic residues in the secondary binding site. Simultaneous occupancy of both binding pockets synergistically increases membrane affinity of the PX domain. Electrostatic potential calculations suggest that a highly positive potential near the secondary binding site may facilitate the initial adsorption of the domain to the anionic membrane, which is followed by the binding of PtdIns(3,4,5)P(3) to its binding pocket. Collectively, our results suggest that the interaction of the PLD1 PX domain with PtdIns(3,4,5)P(3) and/or PA (or phosphatidylserine) may be an important factor in the spatiotemporal regulation and activation of PLD1.
Collapse
Affiliation(s)
- Robert V Stahelin
- Department of Chemistry, University of Illinois at Chicago, Illinois 60607. USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Phospholipase D catalyses the hydrolysis of the phosphodiester bond of glycerophospholipids to generate phosphatidic acid and a free headgroup. Phospholipase D activities have been detected in simple to complex organisms from viruses and bacteria to yeast, plants, and mammals. Although enzymes with broader selectivity are found in some of the lower organisms, the plant, yeast, and mammalian enzymes are selective for phosphatidylcholine. The two mammalian phospholipase D isoforms are regulated by protein kinases and GTP binding proteins of the ADP-ribosylation and Rho families. Mammalian and yeast phospholipases D are also potently stimulated by phosphatidylinositol 4,5-bisphosphate. This review discusses the identification, characterization, structure, and regulation of phospholipase D. Genetic and pharmacological approaches implicate phospholipase D in a diverse range of cellular processes that include receptor signaling, control of intracellular membrane transport, and reorganization of the actin cytoskeleton. Most ideas about phospholipase D function consider that the phosphatidic acid product is an intracellular lipid messenger. Candidate targets for phospholipase-D-generated phosphatidic acid include phosphatidylinositol 4-phosphate 5-kinases and the raf protein kinase. Phosphatidic acid can also be converted to two other lipid mediators, diacylglycerol and lyso phosphatidic acid. Coordinated activation of these phospholipase-D-dependent pathways likely accounts for the pleitropic roles for these enzymes in many aspects of cell regulation.
Collapse
Affiliation(s)
- Mark McDermott
- Department of Cell and Developmental Biology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 27599-7090, USA
| | | | | |
Collapse
|
25
|
Ahn M, Lee Y, Sim KB, Min DS, Matsumoto Y, Wie MB, Shin YG, Shin T. Increased expression of phospholipase D in the heart with experimental autoimmune myocarditis in Lewis rats. Immunol Invest 2004; 33:95-105. [PMID: 15015836 DOI: 10.1081/imm-120027688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The expression of phospholipase D (PLD) in the hearts of rats with experimental autoimmune myocarditis (EAM) was studied to elucidate the functional role of PLD in the pathogenesis of EAM. Western blot analysis showed that the level of the PLD1 isoform was significantly increased in the hearts of rats with EAM on days 14, 17 and 21 postimmunization (pi) (P < 0.01; control vs EAM at 14 pi, 17 pi and 21 pi). The phenotypes of cells exhibiting increased PLD1 expression were primarily inflammatory cells, including ED1 positive macrophages, in the inflammatory EAM lesions. Some cardiomyocytes also showed increased PLD1 immunoreaction in and around EAM lesions. Some PLD1-positive cells were also positive for proliferating cell nuclear antigen in some cardiomyocytes or terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end-labeling in some macrophages, suggesting that PLD1 positive cells have a capacity for proliferation or apoptosis depending on cell types in the target organ. Thus, it is postulated that increased expression of PLD1 in EAM may support an early inflammatory response in proliferating inflammatory cells, and its expression in cardiomyocytes may help them to survive by activation of survival factors in hearts with EAM.
Collapse
Affiliation(s)
- Meejung Ahn
- Department of Veterinary Medicine, College of Agriculture and Life Sciences, Cheju National University, Jeju, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Choi WS, Hiragun T, Lee JH, Kim YM, Kim HP, Chahdi A, Her E, Han JW, Beaven MA. Activation of RBL-2H3 mast cells is dependent on tyrosine phosphorylation of phospholipase D2 by Fyn and Fgr. Mol Cell Biol 2004; 24:6980-92. [PMID: 15282299 PMCID: PMC479740 DOI: 10.1128/mcb.24.16.6980-6992.2004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Both phospholipase D1 (PLD1) and PLD2 regulate degranulation when RBL-2H3 cells are stimulated via the immunoglobulin E receptor, Fc epsilon RI. However, the activation mechanism for PLD2 is unclear. As reported here, PLD2 but not PLD1 is phosphorylated through the Src kinases, Fyn and Fgr, and this phosphorylation appears to regulate PLD2 activation and degranulation. For example, only hemagglutinin-tagged PLD2 was tyrosine phosphorylated in antigen-stimulated cells that had been made to express HA-PLD1 and HA-PLD2. This phosphorylation was blocked by a Src kinase inhibitor or by small interfering RNAs directed against Fyn and Fgr and was enhanced by overexpression of Fyn and Fgr but not by other Src kinases. The phosphorylation and activity of PLD2 were further enhanced by the tyrosine phosphatase inhibitor, Na(3)VO(4). Mutation of PLD2 at tyrosines 11, 14, 165, or 470 partially impaired, and mutation of all tyrosines blocked, PLD2 phosphorylation and activation, although two of these mutations were detrimental to PLD2 function. PLD2 phosphorylation preceded degranulation, both events were equally sensitive to inhibition of Src kinase activity, and both were enhanced by coexpression of PLD2 and the Src kinases. The findings provide the first description of a mechanism for activation of PLD2 in a physiological setting and of a role for Fgr in Fc epsilon RI-mediated signaling.
Collapse
Affiliation(s)
- Wahn Soo Choi
- Laboratory of Molecular Immunology, National, Heart, Lung, and Blood Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
It has been well documented that protein kinase C (PKC) plays an important role in regulation of phospholipase D (PLD) activity. Although PKC regulation of PLD1 activity has been studied extensively, the role of PKC in PLD2 regulation remains to be established. In the present study it was demonstrated that phorbol 12-myristate 13-acetate (PMA) induced PLD2 activation in COS-7 cells. PLD2 was also phosphorylated on both serine and threonine residues after PMA treatment. PKC inhibitors Ro-31-8220 and bisindolylmaleimide I inhibited both PMA-induced PLD2 phosphorylation and activation. However, Gö 6976, a PKC inhibitor relatively specific for conventional PKC isoforms, almost completely abolished PLD2 phosphorylation by PMA but only slightly inhibited PLD2 activation. Furthermore, time course studies showed that phosphorylation of PLD2 lagged behind its activation by PMA. Concentration curves for PMA action on PLD2 phosphorylation and activation also showed that PLD2 was activated by PMA at concentrations at which PMA didn't induce phosphorylation. A kinase-deficient mutant of PKCalpha stimulated PLD2 activity to an even higher level than wild type PKCalpha. Co-expression of wild type PKCalpha, but not PKCdelta, greatly enhanced both basal and PMA-induced PLD2 phosphorylation. A PKCdelta-specific inhibitor, rottlerin, failed to inhibit PMA-induced PLD2 phosphorylation and activation. Co-immunoprecipitation studies indicated an association between PLD2 and PKCalpha under basal conditions that was further enhanced by PMA. Time course studies of the effects of PKCalpha on PLD2 showed that as the phosphorylation of PLD2 increased, its activity declined. In summary, the data demonstrated that PLD2 is activated and phosphorylated by PMA and PKCalpha in COS-7 cells. However, the phosphorylation is not required for PKCalpha to activate PLD2. It is suggested that interaction rather than phosphorylation underscores the activation of PLD2 by PKC in vivo and that phosphorylation may contribute to the inactivation of the enzyme.
Collapse
Affiliation(s)
- Jun-Song Chen
- Howard Hughes Medical Institute and the Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
28
|
Kim DS, Yoon MS, Kim TW, Han JS. Thyrotropin-releasing hormone increases phospholipase D activity through stimulation of protein kinase C in GH3 cells. Endocrine 2004; 23:33-8. [PMID: 15034194 DOI: 10.1385/endo:23:1:33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2003] [Revised: 11/30/2003] [Accepted: 12/09/2003] [Indexed: 11/11/2022]
Abstract
Activation of phospholipase D was investigated after treatment of GH3 cells with thyrotropin-releasing hormone. Thyrotropin-releasing hormone treatment resulted in both time- and dose-dependent increases of phospholipase D activity, translocation of protein kinase C-alpha and -beta I isozymes from cytosol to membrane within 30 min, and approx 43-fold increase of phosphatidylinositol-specific phospholipase C activity. Intracellular calcium concentration was rapidly increased and diacyglycerol level remained high up to 3 h after the treatment. Pretreatment of the cells with U73122, a potent inhibitor of phosphatidylinositol-specific phospholipase C, inhibited thyrotropin-releasing hormone-induced phospholipase D activation. Protein kinase C activity was down-regulated by pretreatment of the GH3 cells with either protein kinase C inhibitors (RO320432, GF109203X) or preincubation of the cells with phorbol myristrate acetate (500 nM) for 24 h. This treatment largely abolished the thyrotropin-releasing hormone-induced activation of phospholipase D, thus further confirming the involvement of protein kinase C in the activation. These results suggest that thyrotropin-releasing hormone-induced phospholipase D activation may be due to phosphatidylinositol-specific phospholipase C, and activation of protein kinase C isozymes is responsible for this stimulation.
Collapse
Affiliation(s)
- Dong-Sun Kim
- Department of Internal Medicine, College of Medicine, Hanyang University, Sungdong-Gu, Seoul 133-791, Korea
| | | | | | | |
Collapse
|
29
|
Abstract
The origin of eukaryotes is one of the major challenges of evolutionary cell biology. Other than the endosymbiotic origin of mitochondria and chloroplasts, the steps leading to eukaryotic endomembranes and endoskeleton are poorly understood. Ras-family small GTPases are key regulators of cytoskeleton dynamics, vesicular trafficking and nuclear function. They are specific for eukaryotes and their expansion probably traces the evolution of core eukaryote features. The phylogeny of small GTPases suggests that the first endomembranes to evolve during eukaryote evolution had secretory, and not phagocytic, function. Based on the reconstruction of putative roles for ancestral small GTPases, a hypothetical scenario on the origins of the first endomembranes, the nucleus, and phagocytosis is presented.
Collapse
Affiliation(s)
- Gáspár Jékely
- European Molecular Biology Laboratory, Developmental Biology, Meyerhofstrasse 1., 69117 Heidelberg, Germany.
| |
Collapse
|
30
|
Wang JP, Chang LC, Hsu MF, Lin CN. The blockade of formyl peptide-induced respiratory burst by 2',5'-dihydroxy-2-furfurylchalcone involves phospholipase D signaling in neutrophils. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2003; 368:166-74. [PMID: 12928764 DOI: 10.1007/s00210-003-0782-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2003] [Accepted: 06/27/2003] [Indexed: 11/30/2022]
Abstract
The inhibition of formyl-methionyl-leucyl-phenylalanine (fMLP)-induced respiratory burst by 2',5'-dihydroxy-2-furfurylchalcone (DHFC) was investigated in rat neutrophils, and the underlying mechanism of this inhibition was assessed. DHFC concentration-dependently inhibited superoxide anion (O(2)) generation (IC(50) 4.2+/-1.2 microM), reaching a plateau within 5-10 min preincubation time, and inhibited oxygen consumption (IC(50) 6.9+/-1.9 microM) in rat neutrophils. In cell-free systems, DHFC failed to scavenge the generated during dihydroxyfumaric acid auto-oxidation. DHFC was less effective in the inhibition of both phorbol 12-myristate 13-acetate-activated neutrophil particulate NADPH oxidase activity and arachidonic acid-induced NADPH oxidase activation. In rat neutrophils, DHFC did not exert a cAMP-elevating effect, nor did it affect fMLP-induced [Ca(2+)](i) change to a considerable extent. DHFC slightly reduced fMLP-induced phosphatidylinositol 3-kinase (PI3 K) activation but showed moderate inhibition of Akt phosphorylation. fMLP-induced cellular phospholipase D (PLD) activation was markedly inhibited by DHFC (IC(50) 8.9+/-2.0 microM). In addition, DHFC effectively attenuated the membrane association of protein kinase C (PKC)-alpha, ADP-ribosylation factor (ARF) and Rho A in fMLP-stimulated cells. However, DHFC had no effect on the membrane association of ARF and Rho A caused by guanosine 5'-[gamma-thio]triphosphate (GTPgammaS) in cell lysate. fMLP-stimulated protein tyrosine phosphorylation was weakly attenuated by DHFC. DHFC was more efficient in the inhibition of extracellular signal-regulated kinase (ERK) phosphorylation than p38 mitogen-activated protein kinase (MAPK) phosphorylation. Collectively, these results indicate that the suppression of fMLP-induced respiratory burst by DHFC in rat neutrophils is probably mainly attributable to the inhibition of PLD activation, via the blockade of PKC-alpha, ARF and Rho A membrane association.
Collapse
Affiliation(s)
- Jih-Pyang Wang
- Department of Education and Research, Taichung Veterans General Hospital, 407, Taichung, Taiwan, Republic of China.
| | | | | | | |
Collapse
|
31
|
Abstract
In this report we demonstrate that in human adrenocortical carcinoma NCI H295R cells, a model for adrenal glomerulosa cells, PLD was activated both by AngII and protein kinase C (PKC)-activating phorbol 12-myristate 13-acetate (PMA). However, while PMA triggered sustained PLD activation, AngII induced transient PLD activation, in contrast to results in bovine glomerulosa cells in primary culture. Despite the transient effect of AngII on PLD activity, PLD-derived lipid signals were required for maximal AngII-elicited aldosterone secretion. AngII-induced PLD activation was inhibited by PKC inhibitors, but not by tyrosine kinase or calcium/calmodulin-dependent kinase inhibitors or a calmodulin antagonist. Both AngII- and PMA-stimulated PLD activity was enhanced by phosphoinositide 3-kinase (PI3K) inhibitors. Akt, a downstream protein kinase activated by the products of PI3K, was constitutively active in H295R cells, and this activity was blocked by PI3K inhibitors. These results suggested that in H295R adrenocortical carcinoma cells, AngII-induced PLD activation was promoted by PKC and inhibited by the constitutively active PI3K pathway.
Collapse
Affiliation(s)
- Xiangjian Zheng
- Institute of Molecular Medicine and Genetics/CB-2803, Medical College of Georgia, 1120 15th Street, Augusta, GA 30912-2630, USA
| | | |
Collapse
|
32
|
Oka M, Kageshita T, Ono T, Goto A, Kuroki T, Ichihashi M. Protein kinase C alpha associates with phospholipase D1 and enhances basal phospholipase D activity in a protein phosphorylation-independent manner in human melanoma cells. J Invest Dermatol 2003; 121:69-76. [PMID: 12839565 DOI: 10.1046/j.1523-1747.2003.12300.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
It is well known that phospholipase D plays a crucial part in the signal transduction of many types of cells, and is activated by protein kinase C alpha when cells are stimulated. To elucidate the role of phospholipase D in melanoma, the expression of phospholipase D1 and protein kinase C alpha in primary and metastatic lesions of acral lentiginous melanoma and superficial spreading melanoma was investigated using immunohistologic techniques. In addition, the mechanism of regulation of phospholipase D1 by protein kinase C alpha was examined in a human melanoma cell line HM3KO using an adenovirus-mediated gene transfer technique. Both phospholipase D1 and protein kinase C alpha were strongly expressed in primary and metastatic lesions of superficial spreading melanoma. Conversely, in acral lentiginous melanoma lesions, the expression of these two proteins increased dramatically with tumor progression; the expression of both phospholipase D1 and protein kinase C alpha was almost negative in the radial growth phase of primary acral lentiginous melanoma lesions, and increased synchronously in a progression-related manner in advanced acral lentiginous melanoma lesions, including vertical growth phase and metastatic lesions. Immunoprecipitation study showed that phospholipase D1 and protein kinase C alpha are associated physiologically in resting melanoma cells. Further immunoprecipitation study using HM3KO cells after adenovirus-mediated simultaneous overexpression of phospholipase D1 and protein kinase C alpha, or phospholipase D1 and the kinase-negative mutant of protein kinase C alpha revealed that both protein kinase C alpha and the kinase-negative mutant of protein kinase C alpha are associated with phospholipase D1 in melanoma cells in the absence of an external signal. Overexpression of protein kinase C alpha or the kinase-negative mutant of protein kinase C alpha in melanoma cells by the adenovirus vectors resulted in the enhancement of basal phospholipase D activity in a viral concentration-dependent manner. Furthermore, enhanced basal phospholipase D activity increased the in vitro invasive potential of HM3KO cells. These results suggest that upregulation of phospholipase D1 and protein kinase C alpha plays a part in the progression of acral lentiginous melanoma from the radial growth phase to the vertical growth phase. The present results also suggest that protein kinase C alpha associates with phospholipase D1 and enhances basal phospholipase D activity in a protein phosphorylation-independent manner in melanoma cells, which contributes to the cell's high invasive potential.
Collapse
Affiliation(s)
- Masahiro Oka
- Department of Dermatology, Kobe University Graduate School of Medicine, Kobe, Japan.
| | | | | | | | | | | |
Collapse
|
33
|
Ahn SJ, Yoon MS, Hyuk S, Han W, Yoon YD, Han JS, Noh DY. Phospholipase C-protein kinase C mediated phospholipase D activation pathway is involved in tamoxifen induced apoptosis. J Cell Biochem 2003; 89:520-8. [PMID: 12761885 DOI: 10.1002/jcb.10532] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tamoxifen (TAM) is the endocrine therapeutic agent the most widely used in the treatment of breast cancer, and it operates primarily through the induction of apoptosis. In this study, we attempted to elucidate the non-ER mediated mechanism behind TAM treatment, involving the phospholipase C-protein kinase C (PLC-PKC) mediated phospholipase D (PLD) activation pathway, using multimodality methods. In TAM treated MCF7 cells, the PLC and PLD protein and mRNA levels increased. Phosphatidylethanol (PEt) and diacylglycerol (DAG) generation also increased, showing increased activity of PLD and PLCgamma1. Translocation of PKCalpha, from cytosol to membrane, was observed in TAM treated cells. By showing that both PKC and PLC inhibitors could reduce the effects of TAM-induced PLD activation, we confirmed the role of PKC and PLC as upstream regulators of PLD. Finally, we demonstrated that TAM treatment reduced the viability of MCF7 cells and brought about rapid cell death. From these results, we confirmed the hypothesis that TAM induces apoptosis in breast cancer cells, and that the signal transduction pathway, involving PLD, PLC, and PKC, constitutes one of the possible mechanisms underlying the non-ER mediated effects associated with TAM.
Collapse
Affiliation(s)
- Soo-Jung Ahn
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 110-744, Korea
| | | | | | | | | | | | | |
Collapse
|
34
|
Chahdi A, Choi WS, Kim YM, Beaven MA. Mastoparan selectively activates phospholipase D2 in cell membranes. J Biol Chem 2003; 278:12039-45. [PMID: 12556526 DOI: 10.1074/jbc.m212084200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Both known isoforms of phospholipase (PL) D, PLD1 and PLD2, require phosphatidylinositol 4,5-bisphosphate for activity. However, PLD2 is fully active in the presence of this phospholipid, whereas PLD1 activation is dependent on additional factors such as ADP-ribosylation factor-1 (ARF-1) and protein kinase Calpha. We find that mastoparan, an activator of G(i) and mast cells, stimulates an intrinsic PLD activity, most likely PLD2, in fractions enriched in plasma membranes from rat basophilic leukemia 2H3 mast cells. Overexpression of PLD2, but not of PLD1, results in a large increase in the mastoparan-inducible PLD activity in membrane fractions, particularly those enriched in plasma membranes. As in previous studies, expressed PLD2 is localized primarily in the plasma membrane and PLD1 in granule membranes. Studies with pertussis toxin and other agents indicate that mastoparan stimulates PLD2 independently of G(i), ARF-1, protein kinase C, and calcium. Kinetic studies indicate that mastoparan interacts synergistically with phosphatidylinositol 4,5-bisphosphate and that oleate, itself a weak stimulant of PLD2 at low concentrations, is a competitive inhibitor of mastoparan stimulation of PLD2. Therefore, mastoparan may be useful for investigating the regulation of PLD2, particularly in view of the well studied molecular interactions of mastoparan with certain other strategic signaling proteins.
Collapse
Affiliation(s)
- Ahmed Chahdi
- Laboratory of Molecular Immunology, NHLBI, National Institutes of Health, Bethesda, Maryland 20892-1760, USA
| | | | | | | |
Collapse
|
35
|
Chang LC, Chen CM, Wang JP. Inhibition of formyl-methionyl-leucyl-phenylalanine-stimulated phospholipase D activation in rat neutrophils by the synthetic isoquinoline DMDI. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1620:191-8. [PMID: 12595089 DOI: 10.1016/s0304-4165(02)00532-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The expression of phospholipase D (PLD) isoenzymes in neutrophils was investigated using reverse transcription-polymerase chain reaction analysis. Amplification products of predicted size were obtained from rat neutrophils with nucleotide sequences corresponding to PLD1a and PLD2. 1-(3',4'-Dimethoxybenzyl)-6,7-dichloroisoquinoline (DMDI) inhibited the formyl-methionyl-leucyl-phenylalanine (fMLP)-stimulated PLD activation in rat neutrophils. The underlying cellular signaling mechanism of DMDI inhibition was investigated. The fMLP-induced protein tyrosine phosphorylation and the membrane translocation of ADP-ribosylation factor (ARF) and Rho A in neutrophils was attenuated by DMDI in a concentration-dependent manner. However, neither the membrane association of protein kinase C-alpha and -beta isoenzymes in fMLP-stimulated cells nor the GTPgammaS- and phorbol 12-myristate 13-acetate-stimulated membrane translocation of ARF and Rho A in a cell-free system was affected significantly by DMDI. These results indicate that the expression of PLD1a and PLD2 mRNA in neutrophils. Attenuation of protein tyrosine phosphorylation and the membrane association of ARF and Rho A probably play a concerted role in the inhibition of PLD by DMDI in rat neutrophils in response to fMLP.
Collapse
Affiliation(s)
- Ling-Chu Chang
- Department of Education and Research, Taichung Veterans General Hospital, 160, Chung Kang Road, Sec. 3, Taiwan, ROC
| | | | | |
Collapse
|
36
|
Abstract
It has been suggested that protein-protein interaction is important for protein kinase C (PKC) alpha to activate phospholipase D1 (PLD1). To determine the one or more sites on PKCalpha that are involved in binding to PLD1, fragments containing the regulatory domain, catalytic domain, and C1-C3 domain of PKCalpha were constructed and shown to be functional, but they all failed to bind and activate PLD1 in vivo and in vitro. A C-terminal 23-amino acid (aa) deletion mutant of PKCalpha was also found to be inactive. To define the binding/activation site(s) in the C terminus of PKCalpha, 1- to 11-aa deletion mutants were made in this terminus. Deletion of up to 9 aa did not alter the ability of PKCalpha to bind and activate PLDl, whereas a 10-aa deletion was inactive. The residue at position 10 was Phe(663). Mutations of this residue (F663D and F663A) caused loss of binding, activation, and phosphorylation of PLD1, indicating that Phe(663) is essential for these activities. Time course experiments showed that the activation of PLD1 by PMA was much faster than its phosphorylation, and its activity decreased as phosphorylation increased with time. Staurosporine, a PKC inhibitor, completely inhibited PLD1 phosphorylation in response to 4beta-phorbol 12-myristate 13-acetate PMA and blocked the later decrease in PLD activity. The same results were found with the D481E mutant of PKCalpha, which is unable to phosphorylate PLD1. These results indicate that neither the regulatory nor catalytic domains of PKCalpha alone can bind to or activate PLD1 and that a residue in the C terminus of PKCalpha (Phe(663)) is required for these effects. The initial activation of PLD1 by PMA is highly correlated with the binding of PKCalpha. Although PKCalpha can phosphorylate PLD1, this is a relatively slow process and is associated with inactivation of the enzyme.
Collapse
Affiliation(s)
- Tianhui Hu
- Howard Hughes Medical Institute and the Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
37
|
Ryu GR, Kim MJ, Song CH, Sim SS, Min DS, Rhie DJ, Yoon SH, Hahn SJ, Kim MS, Jo YH. Site-Specific Distribution of Phospholipase D Isoforms in the Rat Pancreas. Acta Histochem Cytochem 2003. [DOI: 10.1267/ahc.36.51] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Gyeong Ryul Ryu
- Department of Physiology, College of Medicine, The Catholic University of Korea
| | - Myung-Jun Kim
- Department of Physiology, College of Medicine, The Catholic University of Korea
| | - Chan-Hee Song
- Department of Physiology, College of Medicine, The Catholic University of Korea
| | - Sang Soo Sim
- Department of Pathophysiology, College of Pharmacy, Chung Ang University
| | - Do Sik Min
- Department of Physiology, College of Medicine, The Catholic University of Korea
| | - Duck-Joo Rhie
- Department of Physiology, College of Medicine, The Catholic University of Korea
| | - Shin Hee Yoon
- Department of Physiology, College of Medicine, The Catholic University of Korea
| | - Sang June Hahn
- Department of Physiology, College of Medicine, The Catholic University of Korea
| | - Myung-Suk Kim
- Department of Physiology, College of Medicine, The Catholic University of Korea
| | - Yang-Hyeok Jo
- Department of Physiology, College of Medicine, The Catholic University of Korea
| |
Collapse
|
38
|
Abstract
Accumulating evidence has recognized phospholipase D (PLD) as an important element in signal transduction of cell responses, including proliferation and differentiation, However, its role in pro-apoptotic, anti-apoptotic or pro-survival signaling is not well-understood. Involvement of PLD in these signaling mechanisms is considered to differ depending on the cell type and the extracellular stimulus.
Collapse
Affiliation(s)
- Yoshinori Nozawa
- Department of Environmental Cell Responses, Gifu International Institute of Biotechnology, Mitakecho 2193-128, Kanigun, Gifu 505-0116, Japan.
| |
Collapse
|
39
|
Abstract
Structural studies of plant and bacterial members of the phospholipase D (PLD) superfamily are providing information about the role of the conserved HKD domains in the structure of the catalytic center and the catalytic mechanism of mammalian PLD isozymes (PLD1 and PLD2). Mutagenesis and sequence comparison studies have also defined the presence of pleckstrin homology and phox homology domains in the N-terminus and have demonstrated that a conserved sequence at the C-terminus is required for catalysis. The N- and C-terminal regions of PLD1 also contain interaction sites for protein kinase C, which can directly activate the enzyme through a non-phosphorylating mechanism. Small G proteins of the Rho and ADP-ribosylation factor families also directly regulate the enzyme, with RhoA binding to a sequence in the C-terminus. Certain tyrosine kinases and members of the Ras subfamily of small G proteins can activate the enzyme, but the mechanisms appear to be indirect. The mechanisms by which agonists activate PLD in vivo probably involve multiple pathways.
Collapse
Affiliation(s)
- John H Exton
- Howard Hughes Medical Institute and Vanderbilt University Medical Center, Nashville, TN 38232-0295, USA.
| |
Collapse
|
40
|
Sonoda H, Aoki J, Hiramatsu T, Ishida M, Bandoh K, Nagai Y, Taguchi R, Inoue K, Arai H. A novel phosphatidic acid-selective phospholipase A1 that produces lysophosphatidic acid. J Biol Chem 2002; 277:34254-63. [PMID: 12063250 DOI: 10.1074/jbc.m201659200] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a lipid mediator with diverse biological properties, although its synthetic pathways have not been completely solved. We report the cloning and characterization of a novel phosphatidic acid (PA)-selective phospholipase A(1) (PLA(1)) that produces 2-acyl-LPA. The PLA(1) was identified in the GenBank(TM) data base as a close homologue of phosphatidylserine (PS)-specific PLA(1) (PS-PLA(1)). When expressed in insect Sf9 cells, this enzyme was recovered from the Triton X-100-insoluble fraction and did not show any catalytic activity toward exogenously added phospholipid substrates. However, culture medium obtained from Sf9 cells expressing the enzyme was found to activate EDG7/LPA(3), a cellular receptor for 2-acyl-LPA. The activation of EDG7 was further enhanced when the cells were treated with phorbol ester or a bacterial phospholipase D, suggesting involvement of phospholipase D in the process. In the latter condition, an increased level of LPA, but not other lysophospholipids, was confirmed by mass spectrometry analyses. Expression of the enzyme is observed in several human tissues such as prostate, testis, ovary, pancreas, and especially platelets. These data show that the enzyme is a membrane-associated PA-selective PLA(1) and suggest that it has a role in LPA production.
Collapse
Affiliation(s)
- Hirofumi Sonoda
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bollag WB, Jung E, Calle RA. Mechanism of angiotensin II-induced phospholipase D activation in bovine adrenal glomerulosa cells. Mol Cell Endocrinol 2002; 192:7-16. [PMID: 12088862 DOI: 10.1016/s0303-7207(02)00134-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Based on previous data demonstrating activation of phospholipase D (PLD) in response to angiotensin II (AngII), we have hypothesized a role for PLD in mediating aldosterone secretion from bovine adrenal glomerulosa cells. In this study we demonstrate that a PLD-generated signal(s) is required for the AngII-elicited secretory response, since interfering with lipid second messenger formation using a primary alcohol inhibited AngII-induced aldosterone secretion, but not that elicited by incubation with a hydrophilic cholesterol analog, 22(R)-hydroxycholesterol, which bypasses signaling pathways. Three mechanisms for hormonal activation of PLD have been described in other systems: direct receptor coupling, activation through protein kinase C (PKC) and a combination of these two mechanisms. Our results indicate that the PKC activator, phorbol 12-myristic 13-acetate (PMA), is able to activate PLD, and that receptor engagement is apparently not necessary for PLD activation in response to this agent. Maximal doses of AngII and PMA produced no additive effect on PLD activation, suggesting that these two agents function through a common PKC pathway. This interpretation was confirmed by the ability of a PKC inhibitor, Gö 6976, to inhibit partially AngII-induced PLD activation. Finally, treatment with the calcium ionophores A23187 or ionomycin or the calcium channel agonist BAY K8644 had no effect on PLD activity. Likewise, inhibiting calcium influx with high-dose nitrendipine affected neither basal PLD activity nor that stimulated by AngII. Thus, our results suggest a role for PKC, independent of calcium influx, in mediating AngII-induced PLD activation in glomerulosa cells.
Collapse
Affiliation(s)
- Wendy B Bollag
- Program in Cell Signaling, Department of Medicine, Institute of Molecular Medicine and Genetics, Medical College of Georgia, 1120 15th Street, 30912-2630, Augusta, GA, USA.
| | | | | |
Collapse
|
42
|
Choi WS, Kim YM, Combs C, Frohman MA, Beaven MA. Phospholipases D1 and D2 regulate different phases of exocytosis in mast cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:5682-9. [PMID: 12023367 DOI: 10.4049/jimmunol.168.11.5682] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The rat mast cell line RBL-2H3 contains both phospholipase D (PLD)1 and PLD2. Previous studies with this cell line indicated that expressed PLD1 and PLD2 are both strongly activated by stimulants of secretion. We now show by use of PLDs tagged with enhanced green fluorescent protein that PLD1, which is largely associated with secretory granules, redistributes to the plasma membrane in stimulated cells by processes reminiscent of exocytosis and fusion of granules with the plasma membrane. These processes and secretion of granules are suppressed by expression of a catalytically inactive mutant of PLD1 or by the presence of 50 mM 1-butanol but not tert-butanol, an indication that these events are dependent on the catalytic activity of PLD1. Of note, cholera toxin induces translocation of PLD1-labeled granules to the plasma membrane but not fusion of granules with plasma membrane or secretion. Subsequent stimulation of calcium influx with Ag or thapsigargin leads to rapid redistribution of PLD1 to the plasma membrane and accelerated secretion. Also of note, PLD1 is recycled from plasma membrane back to granules within 4 h of stimulation. PLD2, in contrast, is largely confined to the plasma membrane, but it too participates in the secretory process, because expression of catalytically inactive PLD2 also blocks secretion. These data indicate a two-step process: translocation of granules to the cell periphery, regulated by granule-associated PLD1, and a calcium-dependent fusion of granules with the plasma membrane, regulated by plasma membrane-associated PLD2 and possibly PLD1.
Collapse
Affiliation(s)
- Wahn Soo Choi
- Laboratory of Molecular Immunology and Light Microscopy Core Facility, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
43
|
Affiliation(s)
- J H Exton
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
44
|
Affiliation(s)
- Zhi Xie
- Department of Molecular Physiology and Biophysics, Howard Hughes Medical Institute, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | |
Collapse
|
45
|
Jiang X, Gutowski S, Singer WD, Sternweis PC. Assays and characterization of mammalian phosphatidylinositol 4,5-bisphosphate-sensitive phospholipase D. Methods Enzymol 2002; 345:328-34. [PMID: 11665616 DOI: 10.1016/s0076-6879(02)45026-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Affiliation(s)
- Xuejun Jiang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | |
Collapse
|
46
|
Ahn BH, Rhim H, Kim SY, Sung YM, Lee MY, Choi JY, Wolozin B, Chang JS, Lee YH, Kwon TK, Chung KC, Yoon SH, Hahn SJ, Kim MS, Jo YH, Min DS. alpha-Synuclein interacts with phospholipase D isozymes and inhibits pervanadate-induced phospholipase D activation in human embryonic kidney-293 cells. J Biol Chem 2002; 277:12334-42. [PMID: 11821392 DOI: 10.1074/jbc.m110414200] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
alpha-Synuclein has been implicated in the pathogenesis of many neurodegenerative diseases, including Parkinson's disease and Alzheimer's disease. Although the function of alpha-synuclein remains largely unknown, recent studies have demonstrated that this protein can interact with phospholipids. To address the role of alpha-synuclein in neurodegenerative disease, we have investigated whether it binds phospholipase D (PLD) and affects PLD activity in human embryonic kidney (HEK)-293 cells overexpressing wild type alpha-synuclein or the mutant forms of alpha-synuclein (A53T, A30P) associated with Parkinson's disease. Tyrosine phosphorylation of alpha-synuclein appears to play a modulatory role in the inhibition of PLD, because mutation of Tyr(125) to Phe slightly increases inhibitory effect of alpha-synuclein on PLD activity. Treatment with pervanadate or phorbol myristate acetate inhibits PLD more in HEK 293 cells overexpressing alpha-synuclein than in control cells. Binding of alpha-synuclein to PLD requires phox and pleckstrin homology domain of PLD and the amphipathic repeat region and non-Abeta component of alpha-synuclein. Although biologically important, co-transfection studies indicate that the interaction of alpha-synuclein with PLD does not influence the tendency of alpha-synuclein to form pathological inclusions. These results suggest that the association of alpha-synuclein with PLD, and modulation of PLD activity, is biologically important, but PLD does not appear to play an essential role in the pathophysiology of alpha-synuclein.
Collapse
Affiliation(s)
- Bong-Hyun Ahn
- Department of Physiology, Research Institute of Molecular Genetics, and Anatomy, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lee S, Kim JH, Lee CS, Kim JH, Kim Y, Heo K, Ihara Y, Goshima Y, Suh PG, Ryu SH. Collapsin response mediator protein-2 inhibits neuronal phospholipase D(2) activity by direct interaction. J Biol Chem 2002; 277:6542-9. [PMID: 11741937 DOI: 10.1074/jbc.m108047200] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although the functional significance of neuronal phospholipase D (PLD) is being recognized, little is known about its regulatory role in neuronal cells. To elucidate the regulatory mechanism of neuronal PLD, we investigated PLD(2)-binding neuronal protein from rat brain cytosol. During the fractionation of rat brain cytosol by four-column chromatography, a 62-kDa PLD(2)-interacting protein was detected by PLD(2) overlay assay and identified as collapsin response mediator protein-2 (CRMP-2), which controls neuronal axon guidance and outgrowth. Using bacterially expressed glutathione S-transferase fusion proteins, we found that two regions (amino acids 65-192 (the phagocytic oxidase domain) and 724-825) of PLD(2) and a single region (amino acids 243-300) of CRMP-2 are required for the direct binding of both proteins. A co-immunoprecipitation study in COS-7 cells also showed an in vivo interaction between CRMP-2 and PLD(2). Interestingly, CRMP-2 was found to potently inhibit PLD(2) activity in a concentration-dependent manner (IC(50) = 30 nm). Overexpression studies also showed that CRMP-2 is an in vivo inhibitor of PLD(2) in PC12 cells. Moreover, increasing the concentration of semaphorin 3A, one of the repulsive axon guidance cues, showed that PLD(2) activity can be inhibited in PC12 cells. Immunocytochemistry further revealed that PLD(2) is co-localized with CRMP-2 in the distal tips of neurites, its possible action site, in differentiated PC12 cells. Taken together, our results indicate that CRMP-2 may interact directly with and inhibit neuronal PLD(2), suggesting that this inhibitory mode of regulation may play a role in neuronal pathfinding during the developmental stage.
Collapse
Affiliation(s)
- Sukmook Lee
- Division of Molecular and Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Dunkirk SG, Wallert MA, Baumgartner ML, Provost JJ. Isolation and characterization of a 66-kDa protein from rat liver plasma membrane with RhoA-stimulated phospholipase D activity. Protein Expr Purif 2002; 24:1-12. [PMID: 11812216 DOI: 10.1006/prep.2001.1541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A 66-kDa molecular weight protein with phospholipase D activity was solubilized and partially purified from rat liver plasma membrane. The activity and regulation of this phospholipase D have been characterized. Immunoblot analyses indicated that the enzyme was distinct from hPLD1 and PLD2, but was recognized by an antibody to the 12 terminal amino acids of PLD1. PLD activity was stimulated by 1-100 microM Ca(2+) and Mg(2+) and displayed a pH optimum of 7.5. Activity was inhibited by both saturated and unsaturated fatty acids. This PLD was activated in an ATP-independent manner by the PKC isozymes alpha and betaII but not activated by other PKC isozymes. It was also stimulated by the small G-proteins RhoA and ARF. RhoA stimulated the greatest activation, followed by ARF and PKC(alpha). This enzyme was further activated in a synergistic manner when combinations of PKC(alpha) and RhoA or ARF were used. This enzyme displayed a greater response activation by RhoA than to activation by ARF. While a potential breakdown product of PLD1, activation by RhoA indicates that the PLD characterized here is distinct from the other PLDs cloned or isolated to date.
Collapse
Affiliation(s)
- Shawn G Dunkirk
- Department of Chemistry and Department of Biology, Minnesota State University Moorhead, Moorhead, Minnesota 56563, USA
| | | | | | | |
Collapse
|
49
|
Sarkar S, Miwa N, Kominami H, Igarashi N, Hayashi S, Okada T, Jahangeer S, Nakamura S. Regulation of mammalian phospholipase D2: interaction with and stimulation by G(M2) activator. Biochem J 2001; 359:599-604. [PMID: 11672434 PMCID: PMC1222181 DOI: 10.1042/0264-6021:3590599] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We have previously reported that a heat-stable activator for ganglioside metabolism, G(M2) activator, potently stimulates ADP-ribosylation factor (ARF)-dependent phospholipase D (PLD) activity (presumably PLD1) in an in vitro system [Nakamura, Akisue, Jinnai, Hitomi, Sarkar, Miwa, Okada, Yoshida, Kuroda, Kikkawa and Nishizuka (1998) Proc. Natl. Acad. Sci. U.S.A. 95, 12249-12253]. However, little is known about the regulation of PLD2. In the present studies we have investigated the regulation of PLD2 by G(M2) activator and various other regulators including ARF. PLD2 was potently stimulated in vitro by G(M2) activator in a time- and dose-dependent manner. Neither ARF nor protein kinase C caused any significant changes in PLD2 activity. Importantly, PLD2 responsiveness to ARF was greatly enhanced by G(M2) activator, suggesting a possible role for G(M2) activator as a coupling factor. G(M2) activator was also demonstrated to physically associate with PLD2 in a stoichiometric manner. Further, PMA stimulation of COS-7 cells overexpressing both G(M2) activator and PLD2 resulted in a marked increase in the association of the two molecules. Interestingly, ARF association with PLD2 was greatly increased by G(M2) activator. Moreover, G(M2) activator enhanced PMA-induced PLD activity in a synergistic manner with ARF in streptolysin-O-permeabilized, cytosol-depleted HL-60 cells, suggesting that G(M2) activator may regulate PLD in a concerted manner with other factors, including ARF, inside the cells.
Collapse
Affiliation(s)
- S Sarkar
- Division of Biochemistry, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Lee S, Park JB, Kim JH, Kim Y, Kim JH, Shin KJ, Lee JS, Ha SH, Suh PG, Ryu SH. Actin Directly Interacts with Phospholipase D, Inhibiting Its Activity. J Biol Chem 2001; 276:28252-60. [PMID: 11373276 DOI: 10.1074/jbc.m008521200] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mammalian phospholipase D (PLD) plays a key role in several signal transduction pathways and is involved in many diverse functions. To elucidate the complex molecular regulation of PLD, we investigated PLD-binding proteins obtained from rat brain extract. Here we report that a 43-kDa protein in the rat brain, beta-actin, acts as a major PLD2 direct-binding protein as revealed by peptide mass fingerprinting in combination with matrix-assisted laser desorption ionization/time-of-flight mass spectrometry. We also determined that the region between amino acids 613 and 723 of PLD2 is required for the direct binding of beta-actin, using bacterially expressed glutathione S-transferase fusion proteins of PLD2 fragments. Intriguingly, purified beta-actin potently inhibited both phosphatidylinositol-4,5-bisphosphate- and oleate-dependent PLD2 activities in a concentration-dependent manner (IC50 = 5 nm). In a previous paper, we reported that alpha-actinin inhibited PLD2 activity in an interaction-dependent and an ADP-ribosylation factor 1 (ARF1)-reversible manner (Park, J. B., Kim, J. H., Kim, Y., Ha, S. H., Kim, J. H., Yoo, J.-S., Du, G., Frohman, M. A., Suh, P.-G., and Ryu, S. H. (2000) J. Biol. Chem. 275, 21295-21301). In vitro binding analyses showed that beta-actin could displace alpha-actinin binding to PLD2, demonstrating independent interaction between cytoskeletal proteins and PLD2. Furthermore, ARF1 could steer the PLD2 activity in a positive direction regardless of the inhibitory effect of beta-actin on PLD2. We also observed that beta-actin regulates PLD1 and PLD2 with similar binding and inhibitory potencies. Immunocytochemical and co-immunoprecipitation studies demonstrated the in vivo interaction between the two PLD isozymes and actin in cells. Taken together, these results suggest that the regulation of PLD by cytoskeletal proteins, beta-actin and alpha-actinin, and ARF1 may play an important role in cytoskeleton-related PLD functions.
Collapse
Affiliation(s)
- S Lee
- Division of Molecular and Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|