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










Publication year range
1.
Chem Phys Lipids ; 235: 105050, 2021 03.
Article in English | MEDLINE | ID: mdl-33422547

ABSTRACT

Phospholipase C (PLC) ß and ε enzymes hydrolyze phosphatidylinositol (PI) lipids in response to direct interactions with heterotrimeric G protein subunits and small GTPases, which are activated downstream of G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs). PI hydrolysis generates second messengers that increase the intracellular Ca2+ concentration and activate protein kinase C (PKC), thereby regulating numerous physiological processes. PLCß and PLCε share a highly conserved core required for lipase activity, but use different strategies and structural elements to autoinhibit basal activity, bind membranes, and engage G protein activators. In this review, we discuss recent structural insights into these enzymes and the implications for how they engage membranes alone or in complex with their G protein regulators.


Subject(s)
Cell Membrane/metabolism , Phosphoinositide Phospholipase C/metabolism , Phospholipase C beta/metabolism , Cell Membrane/chemistry , Humans , Models, Molecular , Phosphoinositide Phospholipase C/chemistry , Phospholipase C beta/chemistry , Protein Conformation
2.
Mol Cell ; 80(6): 933-934, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33338407

ABSTRACT

Pfeil et al., (2020) examine the mechanism of Gi-stimulated Ca2+ release in cells and find an unexpected role for Gαq in Gßγ-dependent activation of phospholipase Cß (PLCß).


Subject(s)
Calcium , GTP-Binding Protein alpha Subunits, Gq-G11 , Calcium/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Phospholipase C beta/genetics , Phospholipase C beta/metabolism , Signal Transduction
3.
J Biol Chem ; 295(49): 16562-16571, 2020 12 04.
Article in English | MEDLINE | ID: mdl-32948655

ABSTRACT

Phospholipase Cε (PLCε) is activated downstream of G protein-coupled receptors and receptor tyrosine kinases through direct interactions with small GTPases, including Rap1A and Ras. Although Ras has been reported to allosterically activate the lipase, it is not known whether Rap1A has the same ability or what its molecular mechanism might be. Rap1A activates PLCε in response to the stimulation of ß-adrenergic receptors, translocating the complex to the perinuclear membrane. Because the C-terminal Ras association (RA2) domain of PLCε was proposed to the primary binding site for Rap1A, we first confirmed using purified proteins that the RA2 domain is indeed essential for activation by Rap1A. However, we also showed that the PLCε pleckstrin homology (PH) domain and first two EF hands (EF1/2) are required for Rap1A activation and identified hydrophobic residues on the surface of the RA2 domain that are also necessary. Small-angle X-ray scattering showed that Rap1A binding induces and stabilizes discrete conformational states in PLCε variants that can be activated by the GTPase. These data, together with the recent structure of a catalytically active fragment of PLCε, provide the first evidence that Rap1A, and by extension Ras, allosterically activate the lipase by promoting and stabilizing interactions between the RA2 domain and the PLCε core.


Subject(s)
Phosphoinositide Phospholipase C/metabolism , rap1 GTP-Binding Proteins/metabolism , Allosteric Regulation , GTP Phosphohydrolases/metabolism , Humans , Models, Molecular , Mutagenesis, Site-Directed , Phosphoinositide Phospholipase C/chemistry , Phosphoinositide Phospholipase C/genetics , Pleckstrin Homology Domains , Protein Binding , Protein Domains , Protein Structure, Tertiary , Protein Transport , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Scattering, Small Angle , X-Ray Diffraction , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/genetics
4.
Commun Biol ; 3(1): 445, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796910

ABSTRACT

Phospholipase Cε (PLCε) generates lipid-derived second messengers at the plasma and perinuclear membranes in the cardiovascular system. It is activated in response to a wide variety of signals, such as those conveyed by Rap1A and Ras, through a mechanism that involves its C-terminal Ras association (RA) domains (RA1 and RA2). However, the complexity and size of PLCε has hindered its structural and functional analysis. Herein, we report the 2.7 Å crystal structure of the minimal fragment of PLCε that retains basal activity. This structure includes the RA1 domain, which forms extensive interactions with other core domains. A conserved amphipathic helix in the autoregulatory X-Y linker of PLCε is also revealed, which we show modulates activity in vitro and in cells. The studies provide the structural framework for the core of this critical cardiovascular enzyme that will allow for a better understanding of its regulation and roles in disease.


Subject(s)
Phosphoinositide Phospholipase C/chemistry , Phosphoinositide Phospholipase C/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Animals , Crystallography, X-Ray , Enzyme Stability , Models, Biological , Mutation/genetics , Protein Domains , Protein Structure, Secondary , Rats , Transition Temperature
6.
Biochemistry ; 58(32): 3454-3467, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31322863

ABSTRACT

Phospholipase Cß (PLCß) enzymes are peripheral membrane proteins required for normal cardiovascular function. PLCß hydrolyzes phosphatidylinositol 4,5-bisphosphate, producing second messengers that increase intracellular Ca2+ level and activate protein kinase C. Under basal conditions, PLCß is autoinhibited by its C-terminal domains and by the X-Y linker, which contains a stretch of conserved acidic residues required for interfacial activation. Following stimulation of G protein-coupled receptors, the heterotrimeric G protein subunit Gαq allosterically activates PLCß and helps orient the activated complex at the membrane for efficient lipid hydrolysis. However, the molecular basis for how the PLCß X-Y linker, its C-terminal domains, Gαq, and the membrane coordinately regulate activity is not well understood. Using compressed lipid monolayers and atomic force microscopy, we found that a highly conserved acidic region of the X-Y linker is sufficient to regulate adsorption. Regulation of adsorption and activity by the X-Y linker also occurs independently of the C-terminal domains. We next investigated whether Gαq-dependent activation of PLCß altered interactions with the model membrane. Gαq increased PLCß adsorption in a manner that was independent of the PLCß regulatory elements and targeted adsorption to specific regions of the monolayer in the absence of the C-terminal domains. Thus, the mechanism of Gαq-dependent activation likely includes a spatial component.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Lipids/chemistry , Phospholipase C beta/metabolism , Adsorption , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Models, Molecular , Phospholipase C beta/chemistry , Protein Binding , Protein Conformation
7.
Cell Signal ; 62: 109349, 2019 10.
Article in English | MEDLINE | ID: mdl-31254604

ABSTRACT

Phospholipase Cß (PLCß) enzymes regulate second messenger production following the activation of G protein-coupled receptors (GPCRs). Under basal conditions, these enzymes are maintained in an autoinhibited state by multiple elements, including an insertion within the catalytic domain known as the X-Y linker. Although the PLCß X-Y linker is variable in sequence and length, its C-terminus is conserved and features an acidic stretch, followed by a short helix. This helix interacts with residues near the active site, acting as a lid to sterically prevent substrate binding. However, deletions that remove the acidic stretch of the X-Y linker increase basal activity to the same extent as deletion of the entire X-Y linker. Thus, the acidic stretch may be the linchpin in autoinhibition mediated by the X-Y linker. We used site-directed mutagenesis and biochemical assays to investigate the importance of this acidic charge in mediating PLCß3 autoinhibition. Loss of the acidic charge in the X-Y linker increases basal activity and decreases stability, consistent with loss of autoinhibition. However, introduction of compensatory electrostatic mutations on the surface of the PLCß3 catalytic domain restore activity to basal levels. Thus, intramolecular electrostatics modulate autoinhibition by the X-Y linker.


Subject(s)
Catalytic Domain/genetics , Phospholipase C beta/genetics , Protein Conformation, alpha-Helical , Static Electricity , Humans , Mutagenesis, Site-Directed , Phospholipase C beta/antagonists & inhibitors , Phosphorylation , Receptors, G-Protein-Coupled/genetics
8.
Anal Chem ; 91(13): 8466-8475, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31247720

ABSTRACT

Intracellular pH plays a key role in physiology, and its measurement in living specimens remains a crucial task in biology. Fluorescent protein-based pH sensors have gained widespread use, but there is limited spectral diversity for multicolor detection, and it remains a challenge to measure absolute pH values. Here we demonstrate that mCherryTYG is an excellent fluorescence lifetime pH sensor that significantly expands the modalities available for pH quantification in live cells. We first report the 1.09 Å X-ray crystal structure of mCherryTYG, exhibiting a fully matured chromophore. We next determine that it has an extraordinarily large dynamic range with a 2 ns lifetime change from pH 5.5 to 9.0. Critically, we find that the sensor maintains a p Ka of 6.8 independent of environment, whether as the purified protein in solution or expressed in live cells. Furthermore, the lifetime measurements are robustly independent of total fluorescence intensity and scatter. We demonstrate that mCherryTYG is a highly effective sensor using time-resolved fluorescence spectroscopy on live-cell suspensions, which has been previously overlooked as an easily accessible approach for quantifying intracellular pH. As a red fluorescent sensor, we also demonstrate that mCherryTYG is spectrally compatible with the ATeam sensor and EGFP for simultaneous dual-color measurements of intracellular pH, ATP, and extracellular pH. In a proof-of-concept, we quantify acute respiration-dependent pH homeostasis that exhibits a stoichiometric relationship with the ATP-generating capacity of the carbon fuel choice in E. coli. Broadly speaking, our work presents a previously unemployed methodology that will greatly facilitate continuous pH quantification.


Subject(s)
Biosensing Techniques/methods , Cell Respiration , Escherichia coli/metabolism , Fluorescence , Green Fluorescent Proteins/metabolism , Homeostasis , Spectrometry, Fluorescence/methods , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Hydrogen-Ion Concentration
9.
J Biol Chem ; 294(20): 8148-8160, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30940727

ABSTRACT

Regulator of G protein signaling (RGS) proteins are negative regulators of G protein-coupled receptor (GPCR) signaling through their ability to act as GTPase-activating proteins (GAPs) for activated Gα subunits. Members of the RZ subfamily of RGS proteins bind to activated Gαo, Gαz, and Gαi1-3 proteins in the nervous system and thereby inhibit downstream pathways, including those involved in Ca2+-dependent signaling. In contrast to other RGS proteins, little is known about RZ subfamily structure and regulation. Herein, we present the 1.5-Å crystal structure of RGS17, the most complete and highest-resolution structure of an RZ subfamily member to date. RGS17 cocrystallized with Ca2+ bound to conserved positions on the predicted Gα-binding surface of the protein. Using NMR chemical shift perturbations, we confirmed that Ca2+ binds in solution to the same site. Furthermore, RGS17 had greater than 55-fold higher affinity for Ca2+ than for Mg2+ Finally, we found that Ca2+ promotes interactions between RGS17 and activated Gα and decreases the Km for GTP hydrolysis, potentially by altering the binding mechanism between these proteins. Taken together, these findings suggest that Ca2+ positively regulates RGS17, which may represent a general mechanism by which increased Ca2+ concentration promotes the GAP activity of the RZ subfamily, leading to RZ-mediated inhibition of Ca2+ signaling.


Subject(s)
Calcium Signaling , Calcium/chemistry , RGS Proteins/chemistry , Calcium/metabolism , Crystallography, X-Ray , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/genetics , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , Magnesium/chemistry , Magnesium/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism
10.
J Biol Chem ; 293(45): 17477-17490, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30242131

ABSTRACT

Phospholipase C (PLC) enzymes produce second messengers that increase the intracellular Ca2+ concentration and activate protein kinase C (PKC). These enzymes also share a highly conserved arrangement of core domains. However, the contributions of the individual domains to regulation are poorly understood, particularly in isoforms lacking high-resolution information, such as PLCϵ. Here, we used small-angle X-ray scattering (SAXS), EM, and functional assays to gain insights into the molecular architecture of PLCϵ, revealing that its PH domain is conformationally dynamic and essential for activity. We further demonstrate that the PH domain of PLCß exhibits similar dynamics in solution that are substantially different from its conformation observed in multiple previously reported crystal structures. We propose that this conformational heterogeneity contributes to subfamily-specific differences in activity and regulation by extracellular signals.


Subject(s)
Molecular Dynamics Simulation , Pleckstrin Homology Domains , Type C Phospholipases/chemistry , Animals , Humans , Mutation , Rats , Type C Phospholipases/genetics , Type C Phospholipases/metabolism
11.
J Biol Chem ; 293(17): 6387-6397, 2018 04 27.
Article in English | MEDLINE | ID: mdl-29535186

ABSTRACT

Phospholipase C (PLC) enzymes hydrolyze membrane phosphatidylinositol 4,5 bisphosphate (PIP2) and regulate Ca2+ and protein kinase signaling in virtually all mammalian cell types. Chronic activation of the PLCϵ isoform downstream of G protein-coupled receptors (GPCRs) contributes to the development of cardiac hypertrophy. We have previously shown that PLCϵ-catalyzed hydrolysis of Golgi-associated phosphatidylinositol 4-phosphate (PI4P) in cardiac myocytes depends on G protein ßγ subunits released upon stimulation with endothelin-1. PLCϵ binds and is directly activated by Ras family small GTPases, but whether they directly interact with Gßγ has not been demonstrated. To identify PLCϵ domains that interact with Gßγ, here we designed various single substitutions and truncations of WT PLCϵ and tested them for activation by Gßγ in transfected COS-7 cells. Deletion of only a single domain in PLCϵ was not sufficient to completely block its activation by Gßγ, but blocked activation by Ras. Simultaneous deletion of the C-terminal RA2 domain and the N-terminal CDC25 and cysteine-rich domains completely abrogated PLCϵ activation by Gßγ, but activation by the GTPase Rho was retained. In vitro reconstitution experiments further revealed that purified Gßγ directly interacts with a purified fragment of PLCϵ (PLCϵ-PH-RA2) and increases PIP2 hydrolysis. Deletion of the RA2 domain decreased Gßγ binding and eliminated Gßγ stimulation of PIP2 hydrolysis. These results provide first evidence that Gßγ directly interacts with PLCϵ and yield insights into the mechanism by which ßγ subunits activate PLCϵ.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Golgi Apparatus/enzymology , Myocytes, Cardiac/enzymology , Phosphoinositide Phospholipase C/metabolism , cdc25 Phosphatases/metabolism , Animals , COS Cells , Chlorocebus aethiops , Endothelin-1/genetics , Endothelin-1/metabolism , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein gamma Subunits/genetics , Golgi Apparatus/genetics , Myocytes, Cardiac/cytology , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/metabolism , Phosphoinositide Phospholipase C/genetics , Protein Domains , Rats , cdc25 Phosphatases/genetics , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
12.
J Med Genet ; 55(2): 122-130, 2018 02.
Article in English | MEDLINE | ID: mdl-29122926

ABSTRACT

BACKGROUND: Bone dysplasias are a large group of disorders affecting the growth and structure of the skeletal system. METHODS: In the present study, we report the clinical and molecular delineation of a new form of syndromic autosomal recessive spondylometaphyseal dysplasia (SMD) in two Emirati first cousins. They displayed postnatal growth deficiency causing profound limb shortening with proximal and distal segments involvement, narrow chest, radiological abnormalities involving the spine, pelvis and metaphyses, corneal clouding and intellectual disability. Whole genome homozygosity mapping localised the genetic cause to 11q12.1-q13.1, a region spanning 19.32 Mb with ~490 genes. Using whole exome sequencing, we identified four novel homozygous variants within the shared block of homozygosity. Pathogenic variants in genes involved in phospholipid metabolism, such as PLCB4 and PCYT1A, are known to cause bone dysplasia with or without eye anomalies, which led us to select PLCB3 as a strong candidate. This gene encodes phospholipase C ß 3, an enzyme that converts phosphatidylinositol 4,5 bisphosphate (PIP2) to inositol 1,4,5 triphosphate (IP3) and diacylglycerol. RESULTS: The identified variant (c.2632G>T) substitutes a serine for a highly conserved alanine within the Ha2' element of the proximal C-terminal domain. This disrupts binding of the Ha2' element to the catalytic core and destabilises PLCB3. Here we show that this hypomorphic variant leads to elevated levels of PIP2 in patient fibroblasts, causing disorganisation of the F-actin cytoskeleton. CONCLUSIONS: Our results connect a homozygous loss of function variant in PLCB3 with a new SMD associated with corneal dystrophy and developmental delay (SMDCD).


Subject(s)
Corneal Dystrophies, Hereditary/genetics , Osteochondrodysplasias/genetics , Phosphatidylinositols/metabolism , Phospholipase C beta/genetics , Amino Acid Substitution , Child , Child, Preschool , Chromosomes, Human, Pair 11 , Corneal Dystrophies, Hereditary/etiology , Developmental Disabilities/etiology , Developmental Disabilities/genetics , Female , Homozygote , Humans , Infant, Newborn , Intellectual Disability/genetics , Male , Osteochondrodysplasias/etiology , Pedigree , Phosphatidylinositols/genetics , Phospholipase C beta/metabolism , Signal Transduction/genetics
13.
Biochemistry ; 56(41): 5604-5614, 2017 10 17.
Article in English | MEDLINE | ID: mdl-28945350

ABSTRACT

Phospholipase Cß (PLCß) enzymes hydrolyze phosphatidylinositol 4,5-bisphosphate to produce second messengers that regulate intracellular Ca2+, cell proliferation, and survival. Their activity is dependent upon interfacial activation that occurs upon localization to cell membranes. However, the molecular basis for how these enzymes productively interact with the membrane is poorly understood. Herein, atomic force microscopy demonstrates that the ∼300-residue C-terminal domain promotes adsorption to monolayers and is required for spatial organization of the protein on the monolayer surface. PLCß variants lacking this C-terminal domain display differences in their distribution on the surface. In addition, a previously identified autoinhibitory helix that binds to the PLCß catalytic core negatively impacts membrane binding, providing an additional level of regulation for membrane adsorption. Lastly, defects in phosphatidylinositol 4,5-bisphosphate hydrolysis also alter monolayer adsorption, reflecting a role for the active site in this process. Together, these findings support a model in which multiple elements of PLCß modulate adsorption, distribution, and catalysis at the cell membrane.


Subject(s)
Lipid Bilayers/metabolism , Models, Molecular , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C beta/metabolism , Adsorption , Amino Acid Substitution , Catalytic Domain , Enzyme Activation , Enzyme Stability , Fluorometry , Gene Deletion , Humans , Hydrolysis , Lipid Bilayers/chemistry , Liposomes , Microscopy, Atomic Force , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phospholipase C beta/chemistry , Phospholipase C beta/genetics , Point Mutation , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
14.
Structure ; 22(12): 1844-1854, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25435326

ABSTRACT

Phospholipase C ß (PLCß) enzymes are dramatically activated by heterotrimeric G proteins. Central to this response is the robust autoinhibition of PLCß by the X-Y linker region within its catalytic core and by the Hα2' helix in the C-terminal extension of the enzyme. The molecular mechanism of each and their mutual dependence are poorly understood. Herein, it is shown that distinct regions within the X-Y linker have specific roles in regulating activity. Most important,an acidic stretch within the linker stabilizes a lid that occludes the active site, consistent with crystal structures of variants lacking this region. Inhibition by the Hα2' helix is independent of the X-Y linker and likely regulates activity by limiting membrane interaction of the catalytic core. Full activation of PLCß thus requires multiple independent molecular events induced by membrane association of the catalytic core and by the binding of regulatory proteins.


Subject(s)
Models, Molecular , Phospholipase C beta/metabolism , Crystallography, X-Ray , Humans , Protein Binding , Protein Conformation
15.
Trends Pharmacol Sci ; 35(1): 23-30, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24287282

ABSTRACT

The heterotrimeric G protein Gαq is a central player in signal transduction, relaying signals from activated G-protein-coupled receptors (GPCRs) to effectors and other proteins to elicit changes in intracellular Ca(2+), the actin cytoskeleton, and gene transcription. Gαq functions at the intracellular surface of the plasma membrane, as do its best-characterized targets, phospholipase C-ß, p63RhoGEF, and GPCR kinase 2 (GRK2). Recent insights into the structure and function of these signaling complexes reveal several recurring themes, including complex multivalent interactions between Gαq, its protein target, and the membrane, that are likely essential for allosteric control and maximum efficiency in signal transduction. Thus, the plasma membrane is not only a source of substrates but also a key player in the scaffolding of Gαq-dependent signaling pathways.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Humans , Models, Molecular , Signal Transduction , Structure-Activity Relationship
16.
Mol Pharmacol ; 84(4): 488-500, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23880553

ABSTRACT

Phospholipase C (PLC) enzymes convert phosphatidylinositol-4,5-bisphosphate into the second messengers diacylglycerol and inositol-1,4,5-triphosphate. The production of these molecules promotes the release of intracellular calcium and activation of protein kinase C, which results in profound cellular changes. The PLCß subfamily is of particular interest given its prominent role in cardiovascular and neuronal signaling and its regulation by G protein-coupled receptors, as PLCß is the canonical downstream target of the heterotrimeric G protein Gαq. However, this is not the only mechanism regulating PLCß activity. Extensive structural and biochemical evidence has revealed regulatory roles for autoinhibitory elements within PLCß, Gßγ, small molecular weight G proteins, and the lipid membrane itself. Such complex regulation highlights the central role that this enzyme plays in cell signaling. A better understanding of the molecular mechanisms underlying the control of its activity will greatly facilitate the search for selective small molecule modulators of PLCß.


Subject(s)
Phospholipase C beta/chemistry , Phospholipase C beta/physiology , Animals , Humans , Isoenzymes/chemistry , Isoenzymes/physiology , Protein Isoforms/chemistry , Protein Isoforms/physiology
17.
Nat Struct Mol Biol ; 20(3): 355-62, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23377541

ABSTRACT

Phospholipase C-ß (PLCß) is directly activated by Gαq, but the molecular basis for how its distal C-terminal domain (CTD) contributes to maximal activity is poorly understood. Herein we present both the crystal structure and cryo-EM three-dimensional reconstructions of human full-length PLCß3 in complex with mouse Gαq. The distal CTD forms an extended monomeric helical bundle consisting of three antiparallel segments with structural similarity to membrane-binding bin-amphiphysin-Rvs (BAR) domains. Sequence conservation of the distal CTD suggests putative membrane and protein interaction sites, the latter of which bind the N-terminal helix of Gαq in both the crystal structure and cryo-EM reconstructions. Functional analysis suggests that the distal CTD has roles in membrane targeting and in optimizing the orientation of the catalytic core at the membrane for maximal rates of lipid hydrolysis.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Phospholipase C beta/chemistry , Animals , Catalytic Domain , Cryoelectron Microscopy , Crystallography, X-Ray , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Mice , Models, Molecular , Phospholipase C beta/metabolism , Protein Conformation , Protein Structure, Tertiary
18.
Nat Struct Mol Biol ; 18(9): 999-1005, 2011 Aug 07.
Article in English | MEDLINE | ID: mdl-21822282

ABSTRACT

The enzyme phospholipase C-ß (PLCß) is a crucial regulator of intracellular calcium levels whose activity is controlled by heptahelical receptors that couple to members of the Gq family of heterotrimeric G proteins. We have determined atomic structures of two invertebrate homologs of PLCß (PLC21) from cephalopod retina and identified a helix from the C-terminal regulatory region that interacts with a conserved surface of the catalytic core of the enzyme. Mutations designed to disrupt the analogous interaction in human PLCß3 considerably increase basal activity and diminish stimulation by Gαq. Gαq binding requires displacement of the autoinhibitory helix from the catalytic core, thus providing an allosteric mechanism for activation of PLCß.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Loligo/enzymology , Phospholipase C beta/chemistry , Sepia/enzymology , Animals , Crystallography, X-Ray , Models, Molecular , Mutagenesis, Site-Directed , Phospholipase C beta/physiology , Protein Structure, Secondary/physiology , Protein Structure, Tertiary
19.
Biochemistry ; 48(51): 12202-12, 2009 Dec 29.
Article in English | MEDLINE | ID: mdl-19919093

ABSTRACT

The RNA recognition motif (or RRM) is a ubiquitous RNA-binding module present in approximately 2% of the proteins encoded in the human genome. This work characterizes an expanded RRM, which is present in the Drosophila Bruno protein, and targets regulatory elements in the oskar mRNA through which Bruno controls translation. In this Bruno RRM, the deletion of 40 amino acids prior to the N-terminus of the canonical RRM resulted in a significantly decreased affinity of the protein for its RNA target. NMR spectroscopy showed that the expanded Bruno RRM contains the familiar RRM fold of four antiparallel beta-strands and two alpha-helices, preceded by a 10-residue loop that contacts helix alpha(1) and strand beta(2); additional amino acids at the N-terminus of the domain are relatively flexible in solution. NMR results also showed that a truncated form of the Bruno RRM, lacking the flexible N-terminal amino acids, forms a stable and complete canonical RRM, so that the loss of RNA binding activity cannot be attributed to disruption of the RRM fold. This expanded Bruno RRM provides a new example of the features that are important for RNA recognition by an RRM-containing protein.


Subject(s)
Drosophila Proteins/chemistry , Drosophila/chemistry , RNA-Binding Proteins/chemistry , RNA/chemistry , Amino Acid Sequence , Animals , Dogs , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Humans , Molecular Sequence Data , Protein Binding , Protein Folding , Protein Structure, Secondary , RNA/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sequence Alignment , Zebrafish
20.
Plant Physiol ; 143(4): 1504-18, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17322339

ABSTRACT

Eukaryotic translation initiation factor-4E (eIF4E) recognizes and binds the m(7) guanosine nucleotide at the 5' end of eukaryotic messenger RNAs; this protein-RNA interaction is an essential step in the initiation of protein synthesis. The structure of eIF4E from wheat (Triticum aestivum) was investigated using a combination of x-ray crystallography and nuclear magnetic resonance (NMR) methods. The overall fold of the crystallized protein was similar to eIF4E from other species, with eight beta-strands, three alpha-helices, and three extended loops. Surprisingly, the wild-type protein did not crystallize with m(7)GTP in its binding site, despite the ligand being present in solution; conformational changes in the cap-binding loops created a large cavity at the usual cap-binding site. The eIF4E crystallized in a dimeric form with one of the cap-binding loops of one monomer inserted into the cavity of the other. The protein also contained an intramolecular disulfide bridge between two cysteines (Cys) that are conserved only in plants. A Cys-to-serine mutant of wheat eIF4E, which lacked the ability to form the disulfide, crystallized with m(7)GDP in its binding pocket, with a structure similar to that of the eIF4E-cap complex of other species. NMR spectroscopy was used to show that the Cys that form the disulfide in the crystal are reduced in solution but can be induced to form the disulfide under oxidizing conditions. The observation that the disulfide-forming Cys are conserved in plants raises the possibility that their oxidation state may have a role in regulating protein function. NMR provided evidence that in oxidized eIF4E, the loop that is open in the ligand-free crystal dimer is relatively flexible in solution. An NMR-based binding assay showed that the reduced wheat eIF4E, the oxidized form with the disulfide, and the Cys-to-serine mutant protein each bind m(7)GTP in a similar and labile manner, with dissociation rates in the range of 20 to 100 s(-1).


Subject(s)
Disulfides/chemistry , Eukaryotic Initiation Factor-4E/chemistry , Triticum/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...