Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 288(45): 32160-32171, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24062308

ABSTRACT

Vesicular monoamine transporter 2 (VMAT2) transports monoamines into storage vesicles in a process that involves exchange of the charged monoamine with two protons. VMAT2 is a member of the DHA12 family of multidrug transporters that belongs to the major facilitator superfamily of secondary transporters. Tetrabenazine (TBZ) is a non-competitive inhibitor of VMAT2 that is used in the treatment of hyperkinetic disorders associated with Huntington disease and Tourette syndrome. Previous biochemical studies suggested that the recognition site for TBZ and monoamines is different. However, the precise mechanism of TBZ interaction with VMAT2 remains unknown. Here we used a random mutagenesis approach and selected TBZ-resistant mutants. The mutations clustered around the lumenal opening of the transporter and mapped to either conserved proline or glycine, or to residues immediately adjacent to conserved proline and glycine. Directed mutagenesis provides further support for the essential role of the latter residues. Our data strongly suggest that the conserved α-helix breaking residues identified in this work play an important role in conformational rearrangements required for TBZ binding and substrate transport. Our results provide a novel insight into the mechanism of transport and TBZ binding by VMAT2.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Tetrabenazine/pharmacology , Vesicular Monoamine Transport Proteins/antagonists & inhibitors , Vesicular Monoamine Transport Proteins/metabolism , Adrenergic Uptake Inhibitors/pharmacokinetics , Animals , Binding Sites , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , HEK293 Cells , Humans , Huntington Disease/drug therapy , Huntington Disease/genetics , Huntington Disease/metabolism , Mutagenesis, Site-Directed , Protein Structure, Secondary , Rats , Saccharomyces cerevisiae , Tetrabenazine/pharmacokinetics , Tourette Syndrome/drug therapy , Tourette Syndrome/genetics , Tourette Syndrome/metabolism , Vesicular Monoamine Transport Proteins/genetics
2.
J Biol Chem ; 283(32): 22257-71, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18505730

ABSTRACT

Rac plays a pivotal role in the assembly of the superoxide-generating NADPH oxidase of phagocytes. In resting cells, Rac is found in the cytosol in complex with Rho GDP dissociation inhibitor (RhoGDI). NADPH oxidase assembly involves dissociation of the Rac.RhoGDI complex and translocation of Rac to the membrane. We reported that liposomes containing high concentrations of monovalent anionic phospholipids cause Rac.RhoGDI complex dissociation ( Ugolev, Y., Molshanski-Mor, S., Weinbaum, C., and Pick, E. (2006) J. Biol. Chem. 281, 19204-19219 ). We now designed an in vitro model mimicking membrane phospholipid remodeling during phagocyte stimulation in vivo. We showed that liposomes of "resting cell membrane" composition (less than 20 mol % monovalent anionic phospholipids), supplemented with 1 mol % of polyvalent anionic phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P(3)) in conjunction with constitutively active forms of the guanine nucleotide exchange factors (GEFs) for Rac, Trio, or Tiam1 and a non-hydrolyzable GTP analogue, cause dissociation of Rac1(GDP).RhoGDI complexes, GDP to GTP exchange on Rac1, and binding of Rac1(GTP) to the liposomes. Complexes were not dissociated in the absence of GEF and GTP, and optimal dissociation required the presence of PtdIns(3,4,5)P(3) in the liposomes. Dissociation of Rac1(GDP).RhoGDI complexes was correlated with the affinity of particular GEF constructs, via the N-terminal pleckstrin homology domain, for PtdIns(3,4,5)P(3) and involved GEF-mediated GDP to GTP exchange on Rac1. Phagocyte membranes enriched in PtdIns(3,4,5)P(3) responded by NADPH oxidase activation upon exposure in vitro to Rac1(GDP).RhoGDI complexes, p67(phox), GTP, and Rac GEF constructs with affinity for PtdIns(3,4,5)P(3) at a level superior to that of native membranes.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Guanosine Triphosphate/metabolism , Liposomes/metabolism , Neuropeptides/metabolism , Phosphatidylinositol Phosphates/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Blood Proteins/metabolism , Enzyme Activation , Guanylyl Imidodiphosphate/metabolism , Guinea Pigs , Mice , Multiprotein Complexes/metabolism , NADPH Oxidases/metabolism , Phosphatidylcholines/metabolism , Phosphatidylserines/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , rac1 GTP-Binding Protein , rho-Specific Guanine Nucleotide Dissociation Inhibitors
3.
J Biol Chem ; 282(30): 22122-39, 2007 Jul 27.
Article in English | MEDLINE | ID: mdl-17548354

ABSTRACT

The superoxide-generating NADPH oxidase is converted to an active state by the assembly of a membrane-localized cytochrome b(559) with three cytosolic components: p47(phox), p67(phox), and GTPase Rac1 or Rac2. Assembly involves two sets of protein-protein interactions: among cytosolic components and among cytosolic components and cytochrome b(559) within its lipid habitat. We circumvented the need for interactions among cytosolic components by constructing a recombinant tripartite chimera (trimera) consisting of the Phox homology (PX) and Src homology 3 (SH3) domains of p47(phox), the tetratricopeptide repeat and activation domains of p67(phox), and full-length Rac1. Upon addition to phagocyte membrane, the trimera was capable of oxidase activation in vitro in the presence of an anionic amphiphile. The trimera had a higher affinity (lower EC(50)) for and formed a more stable complex (longer half-life) with cytochrome b(559) compared with the combined individual components, full-length or truncated. Supplementation of membrane with anionic but not neutral phospholipids made activation by the trimera amphiphile-independent. Mutagenesis, truncations, and domain replacements revealed that oxidase activation by the trimera was dependent on the following interactions: 1) interaction with anionic membrane phospholipids via the poly-basic stretch at the C terminus of the Rac1 segment; 2) interaction with p22(phox) via Trp(193) in the N-terminal SH3 domain of the p47(phox) segment, supplementing the electrostatic attraction; and 3) an intrachimeric bond among the p67(phox) and Rac1 segments complementary to their physical fusion. The PX domain of the p47(phox) segment and the insert domain of the Rac1 segment made only minor contributions to oxidase assembly.


Subject(s)
Cytosol/enzymology , Mutant Chimeric Proteins/metabolism , NADPH Oxidases/metabolism , Phagocytes/physiology , Phospholipids/physiology , Phosphoproteins/metabolism , Recombinant Proteins/metabolism , Superoxides/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Guanylyl Imidodiphosphate/pharmacology , Guinea Pigs , Macromolecular Substances/metabolism , Membrane Lipids/physiology , Mutant Chimeric Proteins/isolation & purification
4.
Methods Mol Biol ; 412: 385-428, 2007.
Article in English | MEDLINE | ID: mdl-18453125

ABSTRACT

The superoxide (O2-)-generating enzyme complex of phagocytes, known as the NADPH oxidase, can be assayed in a number of in vitro cell-free (or broken cell) systems. These consist of a mixture of the individual components of the NADPH oxidase, derived from resting phagocytes or in the form of purified recombinant proteins, exposed to an activating agent (or situation), in the presence of NADPH and oxygen. O2- produced by the mixture is measured by being trapped immediately after its generation with an appropriate acceptor in a kinetic assay, which permits the calculation of the linear rate of O2- production over time. Cell-free assays are distinguished from whole-cell assays or assays performed on membranes derived from stimulated cells by the fact that all components in the reaction are derived from resting, nonstimulated cells and, thus, the steps of NADPH oxidase activation (precatalytic [assembly] and catalytic) occur in vitro. Cell-free assays played a paramount role in the identification of the components of the NADPH oxidase complex, the diagnosis of various forms of chronic granulomatous disease (CGD), and, more recently, the analysis of the domains present on the components of the NADPH oxidase participating in protein-protein interactions leading to the assembly of the active complex.


Subject(s)
Multiprotein Complexes/metabolism , NADPH Oxidases/analysis , Animals , Catalysis , Cell Fractionation , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell-Free System , Cytosol/chemistry , Disposable Equipment , Humans , NADPH Oxidases/metabolism , Phagocytes/enzymology , Protein Binding , Sensitivity and Specificity
5.
J Biol Chem ; 281(28): 19204-19, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16702219

ABSTRACT

Activation of the phagocyte NADPH oxidase involves the assembly of a membrane-localized cytochrome b559 with the cytosolic components p47(phox), p67(phox), p40(phox), and the GTPase Rac (1 or 2). In resting phagocytes, Rac is found in the cytosol as a prenylated protein in the GDP-bound form, associated with the Rho GDP dissociation inhibitor (RhoGDI). In the process of NADPH oxidase activation, Rac is dissociated from RhoGDI and translocates to the membrane, in concert with the other cytosolic components. The mechanism responsible for dissociation of Rac from RhoGDI is poorly understood. We generated Rac(1 or 2) x RhoGDI complexes in vitro from recombinant Rac(1 or 2), prenylated enzymatically, and recombinant RhoGDI, and purified these by anion exchange chromatography. Exposing Rac(1 or 2)(GDP) x RhoGDI complexes to liposomes containing four different anionic phospholipids caused the dissociation of Rac(1 or 2)(GDP) from RhoGDI and its binding to the anionic liposomes. Rac2(GDP) x RhoGDI complexes were more resistant to dissociation, reflecting the lesser positive charge of Rac2. Liposomes consisting of neutral phospholipid did not cause dissociation of Rac(1 or 2) x RhoGDI complexes. Rac1 exchanged to the hydrolysis-resistant GTP analogue, GMPPNP, associated with RhoGDI with lower affinity than Rac1(GDP) and Rac1(GMPPNP) x RhoGDI complexes were more readily dissociated by anionic liposomes. Rac1(GMPPNP) x RhoGDI complexes elicited NADPH oxidase activation in native phagocyte membrane liposomes in the presence of p67(phox), without the need for an anionic amphiphile, as activator. Both Rac1(GDP) x RhoGDI and Rac1(GMPPNP) x RhoGDI complexes elicited amphiphile-independent, p67(phox)-dependent NADPH oxidase activation in phagocyte membrane liposomes enriched in anionic phospholipids but not in membrane liposomes enriched in neutral phospholipids.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/chemistry , Liposomes/chemistry , NADPH Oxidases/chemistry , Phospholipids/chemistry , rac GTP-Binding Proteins/chemistry , rac1 GTP-Binding Protein/chemistry , Animals , Anions , Cytochrome b Group/chemistry , Enzyme Activation , Guinea Pigs , Insecta , Phagocytes/metabolism , Phosphoproteins/metabolism , Photosystem II Protein Complex/chemistry , Protein Transport , rho-Specific Guanine Nucleotide Dissociation Inhibitors , RAC2 GTP-Binding Protein
6.
J Leukoc Biol ; 79(5): 881-95, 2006 May.
Article in English | MEDLINE | ID: mdl-16641134

ABSTRACT

Phagocytes generate superoxide (O2*-) by an enzyme complex known as reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Its catalytic component, responsible for the NADPH-driven reduction of oxygen to O2*-, is flavocytochrome b559, located in the membrane and consisting of gp91phox and p22phox subunits. NADPH oxidase activation is initiated by the translocation to the membrane of the cytosolic components p47phox, p67phox, and the GTPase Rac. Cytochrome b559 is converted to an active form by the interaction of gp91phox with p67phox, leading to a conformational change in gp91phox and the induction of electron flow. We designed a new family of NADPH oxidase activators, represented by chimeras comprising various segments of p67phox and Rac1. The prototype chimera p67phox (1-212)-Rac1 (1-192) is a potent activator in a cell-free system, also containing membrane p47phox and an anionic amphiphile. Chimeras behave like bona fide GTPases and can be prenylated, and prenylated (p67phox -Rac1) chimeras activate the oxidase in the absence of p47phox and amphiphile. Experiments involving truncations, mutagenesis, and supplementation with Rac1 demonstrated that the presence of intrachimeric bonds between the p67phox and Rac1 moieties is an absolute requirement for the ability to activate the oxidase. The presence or absence of intrachimeric bonds has a major impact on the conformation of the chimeras, as demonstrated by fluorescence resonance energy transfer, small angle X-ray scattering, and gel filtration. Based on this, a "propagated wave" model of NADPH oxidase activation is proposed in which a conformational change initiated in Rac is propagated to p67phox and from p67phox to gp91phox.


Subject(s)
Enzyme Reactivators/metabolism , NADPH Oxidases/metabolism , Phagocytes/metabolism , Recombinant Fusion Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cytosol/enzymology , Enzyme Activation/physiology , Enzyme Reactivators/chemical synthesis , Humans , Models, Biological , NADPH Oxidases/genetics , Phagocytes/enzymology , Protein Conformation , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/genetics , Structure-Activity Relationship , rac1 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...