Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Methods Enzymol ; 594: 243-264, 2017.
Article in English | MEDLINE | ID: mdl-28779842

ABSTRACT

Structure determination of G protein-coupled receptors (GPCRs) in the inactive state bound to high-affinity antagonists has been very successful through the implementation of a number of protein engineering and crystallization strategies. However, the structure determination of GPCRs in their fully active state coupled to a G protein is still very challenging. Recently, mini-G proteins were developed, which recapitulate the coupling of a full heterotrimeric G protein to a GPCR despite being less than one-third of the size. This allowed the structure determination of the agonist-bound adenosine A2A receptor (A2AR) coupled to mini-Gs. Although this is extremely encouraging, A2AR is very stable compared with many other GPCRs, particularly when an agonist is bound. In contrast, the agonist-bound conformation of the human corticotropin-releasing factor receptor is considerably less stable, impeding the formation of good quality crystals for structure determination. We have therefore developed a novel strategy for the thermostabilization of a GPCR-mini-G protein complex. In this chapter, we will describe the theoretical and practical principles of the thermostability assay for stabilizing this complex, discuss its strengths and weaknesses, and show some typical results from the thermostabilization process.


Subject(s)
Biochemistry/methods , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/chemistry , Amphibian Proteins/chemistry , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Iodine Radioisotopes/chemistry , Peptide Hormones/chemistry , Protein Stability , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, Corticotropin-Releasing Hormone/chemistry , Receptors, Corticotropin-Releasing Hormone/metabolism
2.
Curr Protoc Pharmacol ; 69: 2.13.1-2.13.26, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26344213

ABSTRACT

G protein-coupled receptors (GPCRs) are rapidly phosphorylated following agonist occupation in a process that mediates receptor uncoupling from its cognate G protein, a process referred to as desensitization. In addition, this process provides a mechanism by which receptors can engage with arrestin adaptor molecules and couple to downstream signaling pathways. The importance of this regulatory process has been highlighted recently by the understanding that ligands can direct receptor signaling along one pathway in preference to another, the phenomenon of signaling bias that is partly mediated by the phosphorylation status or phosphorylation barcode of the receptor. Methods to determine the phosphorylation status of a GPCR in vitro and in vivo are necessary to understand not only the physiological mechanisms involved in GPCR signaling, but also to fully examine the signaling properties of GPCR ligands. This unit describes detailed methods for determining the overall phosphorylation pattern on a receptor (the phosphorylation barcode), as well as mass spectrometry approaches that can define the precise sites that become phosphorylated. These techniques, coupled with the generation and characterization of receptor phosphorylation-specific antibodies, provide a full palate of techniques necessary to determine the phosphorylation status of any given GPCR subtype.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Pharmacology/methods , Phosphopeptides/metabolism , Protein Processing, Post-Translational , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Antibodies, Phospho-Specific/metabolism , Blotting, Western , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Heterotrimeric GTP-Binding Proteins/chemistry , Humans , Ligands , Peptide Mapping , Phosphopeptides/chemistry , Phosphopeptides/genetics , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Tandem Mass Spectrometry
3.
Dev Dyn ; 239(4): 1089-101, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20186915

ABSTRACT

Heterotrimeric G proteins are well known for their roles in signal transduction downstream of G protein-coupled receptors (GPCRs), and both Galpha subunits and tightly associated Gbetagamma subunits regulate downstream effector molecules. Compared to Galpha subunits, the physiological roles of individual Gbeta and Ggamma subunits are poorly understood. In this study, we generated mice deficient in the Gbeta1 gene and found that Gbeta1 is required for neural tube closure, neural progenitor cell proliferation, and neonatal development. About 40% Gbeta1(-/-) embryos developed neural tube defects (NTDs) and abnormal actin organization was observed in the basal side of neuroepithelium. In addition, Gbeta1(-/-) embryos without NTDs showed microencephaly and died within 2 days after birth. GPCR agonist-induced ERK phosphorylation, cell proliferation, and cell spreading, which were all found to be regulated by Galphai and Gbetagamma signaling, were abnormal in Gbeta1(-/-) neural progenitor cells. These data indicate that Gbeta1 is required for normal embryonic neurogenesis.


Subject(s)
Cell Proliferation , Heterotrimeric GTP-Binding Proteins/genetics , Neural Tube Defects/genetics , Neurons/physiology , Stem Cells/physiology , Animals , Brain/abnormalities , Brain/metabolism , Cell Proliferation/drug effects , Down-Regulation/genetics , Embryo, Mammalian , Endothelin-1/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein beta Subunits , Gene Expression Regulation, Developmental , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/metabolism , Lysophospholipids/pharmacology , Mice , Mice, Knockout , Mutagenesis, Insertional , Neural Tube Defects/physiopathology , Neurogenesis/genetics , Neurons/drug effects , Phosphorylation/drug effects , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Stem Cells/drug effects
4.
Tsitologiia ; 50(12): 1036-43, 2008.
Article in Russian | MEDLINE | ID: mdl-19198542

ABSTRACT

The molecular mechanisms of action of the polycationic peptides--polylysine homo- and heterodendrimers on functional activity of biogenic amines- and peptide hormones-sensitive adenylyl; cyclase signaling system (AC system) in the myocardium and the brain of rats were studied. These peptides are expected to be used as highly effective polymer carries for biologically active substances. The polylysine homodendrimers of the third [(NH2)16(Lys)8(Lys)4(Lys)2Lys-Ala-NH2] (I), fourth [(NH2)32(Lys)16(Lys)8(Lys)4(Lys)2Lys-Ala-NH2 (II) and fifth [(NH2)64(Lys)32(Lys)16(Lys)8(Lys)4(Lys)2Lys-Ala-NH2] (III) generations and the polylysine homodendrimers of fifth generation--[(NH2)64(Lys-Glu)32(Lys-Glu)16(Lys-Glu)8(Lys-Glu)4(Lys-Glu)2Lys-Ala-Ala-Lys (ClAc)-Ala-NH2] (IV), [(NH2)64(Lys-Ala)32(Lys-Ala)16(Lys-Ala)8(Lys-Ala)4(Lys-Ala)2Lys-Ala-Lys(ClAc)-Ala-Ala-NH2] (V) and [(NH2)64(Lys-Gly-Gly)32(Lys-Gly-Gly)16(Lys-Gly-Gly)8(Lys-Gly-Gly)4(Lys-Gly-Gly)2 Lys-Gly-Gly-Lys(ClAc)-Ala-Ala-NH2] (VI) showed receptor-independent mechanism of heterotrimeric G-proteins activity, preferably of inhibitory type, interacting with C-terminal regions of their alpha-subunits. The homodendrimers II and III and heterodendrimer V are more effective G-protein activators. The polylysine dendrimers disturbed the functional coupling of the receptors of biogenic amines and peptides hormones with Gi-proteins and, in a lesser extent, Gs-proteins. This is illustrated by the decrease in regulatory effects of the hormones on AX activity and G-protein GTP binding and by the decrease in receptor affinity to agonists in the presence of the polylysine dendrimers, as result of receptor--G-proteins complex dissociation. It was shown also that the molecular mechanisms and the selectivity of the action on the G-proteins of the polylysine dendrimers were similar to those of mastoparan and melittin, natural toxins of insect venom.


Subject(s)
Adenylyl Cyclases/metabolism , Dendrimers/pharmacology , Heterotrimeric GTP-Binding Proteins/drug effects , Polylysine/pharmacology , Receptors, Biogenic Amine/drug effects , Adenylyl Cyclases/drug effects , Animals , Brain/metabolism , Brain/ultrastructure , Cell Membrane/drug effects , Cell Membrane/metabolism , Dendrimers/chemical synthesis , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Myocardium/metabolism , Myocardium/ultrastructure , Peptides/pharmacology , Polylysine/chemical synthesis , Rats , Receptors, Biogenic Amine/agonists , Receptors, Biogenic Amine/antagonists & inhibitors , Secologanin Tryptamine Alkaloids/metabolism , Signal Transduction/drug effects
5.
Circulation ; 113(1): 60-6, 2006 Jan 03.
Article in English | MEDLINE | ID: mdl-16380548

ABSTRACT

BACKGROUND: Diacylglycerol is a lipid second messenger that accumulates in cardiomyocytes when stimulated by Gqalpha protein-coupled receptor (GPCR) agonists such as angiotensin II, phenylephrine, and others. Diacylglycerol functions as a potent activator of protein kinase C (PKC) and is catalyzed by diacylglycerol kinase (DGK) to form phosphatidic acid and inactivated. However, the functional roles of DGK have not been previously examined in the heart. We hypothesized that DGK might prevent GPCR agonist-induced activation of diacylglycerol downstream signaling cascades and subsequent cardiac hypertrophy. METHODS AND RESULTS: To test this hypothesis, we generated transgenic (DGKzeta-TG) mice with cardiac-specific overexpression of DGKzeta. There were no differences in heart size and heart weight between DGKzeta-TG and wild-type littermate mice. The left ventricular function was normal in DGKzeta-TG mice. Continuous administration of subpressor doses of angiotensin II and phenylephrine caused PKC translocation, gene induction of atrial natriuretic factor, and subsequent cardiac hypertrophy in WT mice. However, in DGKzeta-TG mice, neither translocation of PKC nor upregulation of atrial natriuretic factor gene expression was observed after angiotensin II and phenylephrine infusion. Furthermore, in DGKzeta-TG mice, angiotensin II and phenylephrine failed to increase cross-sectional cardiomyocyte areas and heart to body weight ratios. Phenylephrine-induced increases in myocardial diacylglycerol levels were completely blocked in DGKzeta-TG mouse hearts, suggesting that DGKzeta regulated PKC activity by controlling cellular diacylglycerol levels. CONCLUSIONS: These results demonstrated the first evidence that DGKzeta negatively regulated the hypertrophic signaling cascade and resultant cardiac hypertrophy in response to GPCR agonists without detectable adverse effects in in vivo hearts.


Subject(s)
Cardiomegaly/prevention & control , Diacylglycerol Kinase/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11 , Heterotrimeric GTP-Binding Proteins/agonists , Myocardium/metabolism , Angiotensin II/pharmacology , Animals , Diacylglycerol Kinase/genetics , Diglycerides/metabolism , Mice , Mice, Transgenic , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Phenylephrine/pharmacology , Promoter Regions, Genetic , Protein Kinase C/metabolism , RNA, Messenger/analysis , Rats , Signal Transduction/drug effects , Ventricular Myosins/genetics
6.
Br J Pharmacol ; 143(6): 705-14, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15302686

ABSTRACT

The present study investigates the effect of varying ligand structure on the ability of agonists to activate guanine nucleotide-binding proteins of the Gi, Gs and Gq families via the A(1) adenosine receptor. In CHO cells expressing this receptor, inhibition or potentiation of forskolin-stimulated cAMP accumulation was used as an end point to measure the activation of Gi and, in Pertussis toxin (PTX)-treated cells, Gs, respectively. Stimulation of inositol phosphate accumulation in PTX-treated cells was used as an index of Gq activation. CPA (N(6)-cyclopentyladenosine), NECA (5'-N-ethyl-carboxyamidoadenosine) and eight analogues of these ligands presented a range of guanine nucleotide-binding protein (G-protein)-activating profiles. Some ligands could only activate Gi (e.g. 2'deoxyCPA), some primarily Gi and Gs (and only weakly Gq) (e.g. 3'deoxyCPA), highlighting the importance of the ribose hydroxyls in agonist activation of multiple G proteins. CHA (N(6)-cyclohexyladenosine) activated Gi, Gs and Gq, but was more efficacious than CPA in activating Gs. The NECA analogues 5'-N-cyclopropyl-carboxamidoadenosine, 5'-N-cyclobutyl-carboxamidoadenosine and 5'-N-cyclopentyl-carboxamidoadenosine (CPeCA) also activated all three G proteins, although their ability to activate Gs and Gq (relative to CPA) was reduced with increasing substituent size, such that CPeCA produced only a small stimulation (at 100 microM) at Gq, but was a full agonist, relative to CPA, at Gi and Gs. This study suggests that the A(1) adenosine receptor can adopt agonist-specific conformations, arising from small changes in ligand structure, which lead to the differential activation of Gi, Gs and Gq.


Subject(s)
Adenosine A1 Receptor Agonists , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/metabolism , Receptor, Adenosine A1/metabolism , Adenosine-5'-(N-ethylcarboxamide)/analogs & derivatives , Adenosine-5'-(N-ethylcarboxamide)/metabolism , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Humans , Protein Binding/drug effects , Protein Binding/physiology
7.
Methods Enzymol ; 389: 119-30, 2004.
Article in English | MEDLINE | ID: mdl-15313563

ABSTRACT

Analysis of the function of regulator of G-protein signaling (RGS) protein function and their selectivity of action in vivo is complicated by the expression of multiple RGS proteins in a single cell and requires precise control of cytosolic RGS protein concentration. This article describes two experimental systems using pancreatic acinar cells suitable for such analyses. The first is pancreatic acini permeabilized with streptolysin O, which retains agonist responsiveness while allowing RGS proteins and molecules with molecular masses of up to 25-30 kDa access to the cytosol. The second is a whole cell recording of the Ca(2+)-activated Cl- current of single pancreatic acinar cells as a reporter of [Ca2+]i. This system can be used to introduce to the cytosol any protein of interest, including recombinant RGS proteins and RGS protein-scavenging antibodies. The use of these systems to study the specificity of RGS proteins action, the function of their domains, and the role of RGS proteins in controlling Ca2+ oscillations is discussed.


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/metabolism , RGS Proteins/antagonists & inhibitors , RGS Proteins/metabolism , Aniline Compounds , Animals , Bacterial Proteins , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Cells, Cultured , Cytosol/metabolism , Fluorescent Dyes , Humans , Molecular Weight , Pancreas/cytology , Pancreas/drug effects , Pancreas/metabolism , Patch-Clamp Techniques , Protein Structure, Tertiary , RGS Proteins/chemistry , RGS Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Streptolysins/pharmacology , Xanthenes
8.
Br J Pharmacol ; 138(5): 775-86, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12642378

ABSTRACT

(1) The human dopamine D(2long) (D(2L)) receptor was expressed with four different G proteins in Sf9 cells using the baculovirus expression system. When co-expressed with G(i)/G(o) G proteins (G(i1)alpha, G(i2)alpha, G(i3)alpha, or G(o)alpha, plus Gbeta(1) and Ggamma(2)), the receptor displayed a high-affinity binding site for the agonists (dopamine and NPA), which was sensitive to GTP (100 micro M), demonstrating interaction between the receptor and the different G proteins. (2) The receptor to G protein ratio (R : G ratio) was evaluated using [(3)H]-spiperone saturation binding (R) and [(35)S]-GTPgammaS saturation binding (G). R : G ratios of 1 : 12, 1 : 3, 1 : 14 and 1 : 5 were found for G(i1), G(i2), G(i3), and G(o) preparations, respectively. However, when R : G ratios of 1 : 2 and 1 : 12 were compared for G(i2) and G(o), no difference was found for the stimulation of [(35)S]-GTPgammaS binding. (3) Several agonists were tested for their ability to stimulate [(35)S]-GTPgammaS binding to membranes co-expressing the receptor and various G proteins. All the compounds tested showed agonist activity in preparations expressing G(i3) and G(o). However, for G(i2) and G(i1) preparations, compounds such as S-(-)-3-PPP and p-tyramine were unable to stimulate [(35)S]-GTPgammaS binding. (4) Most of the compounds showed higher relative efficacies (compared to dopamine) and higher potencies in the preparation expressing G(o). Comparison of the effects of different agonists in the different preparations showed that each agonist differentially activates the four G proteins. (5) We conclude that the degree of selectivity of G protein activation by the D(2L) receptor can depend on the conformation of the receptor stabilised by an agonist.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/agonists , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/metabolism , Proto-Oncogene Proteins/agonists , Proto-Oncogene Proteins/metabolism , Receptors, Dopamine D2/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Proteins/agonists , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Insecta , Proto-Oncogene Proteins/genetics , Receptors, Dopamine D2/genetics
9.
Planta ; 216(4): 674-85, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12569410

ABSTRACT

Nod factors are lipo-chito-oligosaccharides secreted by rhizobia that initiate many responses in the root hairs of the legume hosts, culminating in deformed hairs. The heterotrimeric G-protein agonists mastoparan, Mas7, melittin, compound 48/80 and cholera toxin provoke root hair deformation, whereas the heterotrimeric G-protein antagonist pertussis toxin inhibits mastoparan and Nod factor NodNGR[S]- (from Rhizobiumsp. NGR234) induced root hair deformation. Another heterotrimeric G-protein antagonist, isotetrandrine, only inhibited root hair deformation provoked by mastoparan and melittin. These results support the notion that G-proteins are implicated in Nod factor signalling. To study the role of G-proteins at a biochemical level, we examined the GTP-binding profiles of root microsomal membrane fractions isolated from the nodulation competent zone of Vigna unguiculata(L.) Walp. GTP competitively bound to the microsomal membrane fractions labelled with [(35)S]GTPgammaS, yielding a two-site displacement curve with displacement constants ( K(i)) of 0.58 micro M and 0.16 mM. Competition with either ATP or GDP revealed a one-site displacement curve with K(i) of 4.4 and 29 micro M, respectively, whereas ADP and UTP were ineffective competitors. The GTP-binding profiles of microsomal membrane fractions isolated from roots pretreated with either NodNGR[S] or the four-sugar, N- N'- N"- N'"-tetracetylchitotetraose (TACT) backbone of Nod factors were significantly altered compared with control microsomal fractions. To identify candidate proteins, membrane proteins were separated by SDS-PAGE and electrotransferred to nitrocellulose. GTP overlay experiments revealed that membrane fractions isolated from roots pretreated with NodNGR[S] or TACT contained two proteins (28 kDa and 25 kDa) with a higher affinity for GTPgammaS than control membrane fractions. Western analysis demonstrated that membranes from the pretreated roots contained more of another protein (~55 kDa) recognised by Galpha(common) antisera. These results provide pharmacological and biochemical evidence supporting the contention that G-proteins are involved in Nod factor signalling and, importantly, implicate monomeric G-proteins in this process.


Subject(s)
Fabaceae/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Lipopolysaccharides/pharmacology , Monomeric GTP-Binding Proteins/metabolism , Rhizobium/growth & development , Binding, Competitive/drug effects , Cholera Toxin/pharmacology , Fabaceae/microbiology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Intercellular Signaling Peptides and Proteins , Lipopolysaccharides/metabolism , Melitten/pharmacology , Monomeric GTP-Binding Proteins/drug effects , Oligosaccharides/metabolism , Oligosaccharides/pharmacology , Peptides , Pertussis Toxin/pharmacology , Plant Roots/drug effects , Plant Roots/growth & development , Plant Roots/metabolism , Rhizobium/chemistry , Signal Transduction/drug effects , Sulfur Radioisotopes , Symbiosis , Wasp Venoms/pharmacology
10.
Hypertension ; 40(5): 660-6, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12411459

ABSTRACT

The Gq-coupled receptor-signaling pathway has been implicated in the cardiac hypertrophic response to stress, but little is actually known about the contributions of Gq signaling in either the heart or the vasculature. Therefore, we developed a line of transgenic mice that express a peptide inhibitor of Gq (GqI) in vascular smooth muscle to determine if vascular Gq signaling was important in the cardiac hypertrophic response. After chronic administration of the Gq agonists phenylephrine, serotonin, and angiotensin II, we observed an attenuation of mean arterial blood pressure and an inhibition of cardiac hypertrophy in the transgenic mice with vascular-specific GqI expression. In contrast, cardiac GqI peptide expression did not attenuate the hypertension or the cardiac hypertrophy. Importantly, all mice were capable of cardiac hypertrophy, because direct beta-adrenergic receptor stimulation induced a similar level of hypertrophy in both lines of transgenic mice. This clearly suggests that after chronic Gq-coupled receptor agonist administration, it is the hypertensive state induced by vascular Gq activation that mediates remodeling of the heart, rather than direct stimulation of cardiac Gq-coupled receptors. Thus, the contribution of vascular Gq-coupled signaling to the development of cardiac hypertrophy is significant and suggests that expression of the GqI peptide is a novel therapeutic strategy to lower Gq-mediated hypertension and cardiac hypertrophy.


Subject(s)
Cardiomegaly/physiopathology , Heart/physiopathology , Heterotrimeric GTP-Binding Proteins/metabolism , Microfilament Proteins , Muscle, Smooth, Vascular/metabolism , Receptors, Cell Surface/agonists , Angiotensin II/pharmacology , Animals , Cardiomegaly/etiology , Catecholamines/blood , Cells, Cultured , Enzyme Activation/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11 , Heart/drug effects , Hemodynamics/drug effects , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/genetics , Infusion Pumps, Implantable , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Muscle Proteins/genetics , Muscle, Smooth, Vascular/drug effects , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Phenylephrine/pharmacology , Receptors, Cell Surface/metabolism , Serotonin/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Time , Vasoconstrictor Agents/pharmacology
11.
Circ Res ; 90(8): 850-7, 2002 May 03.
Article in English | MEDLINE | ID: mdl-11988485

ABSTRACT

Previous attempts to delineate the consequences of Galpha (q) activation in cardiomyocytes relied largely on molecular strategies in cultures or transgenic mice. Modest levels of wild-type Galpha(q) overexpression induce stable cardiac hypertrophy, whereas intense Galpha(q) stimulation induces cardiomyocyte apoptosis. The precise mechanism(s) whereby traditional targets of Galpha (q) subunits that induce hypertrophy also trigger cardiomyocyte apoptosis is not obvious and is explored with recombinant Pasteurella multocida toxin (rPMT, a Galpha(q) agonist). Cells cultured with rPMT display cardiomyocyte enlargement, sarcomeric organization, and increased atrial natriuretic factor expression in association with activation of phospholipase C, novel protein kinase C (PKC) isoforms, extracellular signal-regulated protein kinase (ERK), and (to a lesser extent) JNK/p38-MAPK. rPMT stimulates the ERK cascade via epidermal growth factor (EGF) receptor transactivation in cardiac fibroblasts, but EGF receptor transactivation plays no role in ERK activation in cardiomyocytes. Surprisingly, rPMT (or novel PKC isoform activation by PMA) decreases basal Akt phosphorylation; rPMT prevents Akt phosphorylation by EGF or IGF-1 and functionally augments cardiomyocyte apoptosis in response to H2O2. These results identify a Galpha(q)-PKC pathway that represses basal Akt phosphorylation and impairs Akt stimulation by survival factors. Because inhibition of Akt enhances cardiomyocyte susceptibility to apoptosis, this pathway is predicted to contribute to the transition from hypertrophy to cardiac decompensation and could be targeted for therapy in heart failure.


Subject(s)
Apoptosis , Bacterial Proteins , Bacterial Toxins/pharmacology , Heterotrimeric GTP-Binding Proteins/agonists , Myocardium/metabolism , Protein Serine-Threonine Kinases , Animals , Animals, Newborn , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Cells, Cultured , Drug Synergism , ErbB Receptors/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11 , Hydrogen Peroxide/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Myocardium/cytology , Myocardium/enzymology , Protein Kinase C/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Recombinant Proteins/pharmacology , Signal Transduction , Type C Phospholipases/metabolism
12.
Neuropsychopharmacology ; 26(4): 468-78, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11927171

ABSTRACT

The influence of age, postmortem delay and freezing storage period on receptor-mediated G-protein activity was quantified in cortical membranes from 34 healthy subjects. Concentration-response curves of the [(35)S]GTPgammaS binding stimulation by agonists for alpha(2)-adrenoceptors (UK14304), mu-opioid (DAMGO), 5-HT(1A) (8-OH-DPAT), GABA(B) (baclofen) and muscarinic (carbachol) receptors were analyzed. Immunoreactivities of G(alpha)-protein subunits were also determined. Basal binding and UK14304, 8-OH-DPAT, and baclofen potency to stimulate [(35)S]GTPgammaS binding decreased with aging (1-88 years) without changes of efficacy. DAMGO-mediated stimulation increased both in potency and efficacy with aging. A negative correlation between age and immunoreactivity was observed for G(alphai1/2)-, but not for G(alphai3)-, G(alphao)-,and G(alphas)-proteins. Neither [(35)S]GTPgammaS binding nor G(alpha)-proteins changed with the postmortem delay (8-92 h). Basal [(35)S]GTPgammaS binding decreased with the sample storage period (1-85 months). A careful match between cases and controls should be taken into account when designing signal transduction studies in human disorders, specially for variables such as age and storage period.


Subject(s)
Aging/physiology , Brain Chemistry/physiology , GTP-Binding Proteins/physiology , Postmortem Changes , Tissue Preservation , Adolescent , Adult , Aged , Aging/pathology , Brain/pathology , Child , Child, Preschool , Cryopreservation , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/biosynthesis , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Infant , Membranes/chemistry , Middle Aged , Time Factors
13.
Proc Natl Acad Sci U S A ; 99(6): 3866-71, 2002 Mar 19.
Article in English | MEDLINE | ID: mdl-11891332

ABSTRACT

Signaling via mitogen-activated protein kinases is implicated in heart failure induced by agonists for G protein-coupled receptors that act via the G protein Galphaq. However, this assertion relies heavily on pharmacological inhibitors and dominant-interfering proteins and not on gene deletion. Here, we show that endogenous cardiac MAPK/ERK kinase kinase-1 (MEKK1)/(MAP3K1), a mitogen-activated protein kinase kinase kinase, is activated by heart-restricted overexpression of Galphaq in mice. In cardiac myocytes derived from embryonic stem cells in culture, homozygous disruption of MEKK1 selectively impaired c-Jun N-terminal kinase activity in the absence or presence of phenlyephrine, a Galphaq-dependent agonist. Other terminal mitogen-activated protein kinases were unaffected. In mice, the absence of MEKK1 abolished the increase in cardiac mass, myocyte size, hypertrophy-associated atrial natriuretic factor induction, and c-Jun N-terminal kinase activation by Galphaq, and improved ventricular mechanical function. Thus, MEKK1 mediates cardiac hypertrophy induced by Galphaq in vivo and is a logical target for drug development in heart disease involving this pathway.


Subject(s)
Cardiomegaly/enzymology , Cardiomegaly/physiopathology , Heterotrimeric GTP-Binding Proteins/metabolism , MAP Kinase Kinase Kinase 1 , MAP Kinase Signaling System , Protein Serine-Threonine Kinases/metabolism , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Enzyme Activation , GTP-Binding Protein alpha Subunits, Gq-G11 , Gene Expression , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/genetics , JNK Mitogen-Activated Protein Kinases , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Myocardium/enzymology , Myocardium/metabolism , Protein Serine-Threonine Kinases/genetics , Stem Cells/enzymology , Stem Cells/metabolism
14.
Biochemistry ; 40(35): 10532-41, 2001 Sep 04.
Article in English | MEDLINE | ID: mdl-11523995

ABSTRACT

G protein coupled receptors activate signal transducing guanine nucleotide-binding proteins (G proteins), which consist of an alpha subunit and a betagamma dimer. Whole cell studies have reported that receptors signal through specific betagamma subtypes. Membrane reconstitution studies with the adenosine A(1) and alpha(2A) adrenergic receptors have reached a similar conclusion. We aimed to test the generality of this finding by comparing the gamma subtype specificity for four G(i)-coupled receptors: alpha(2A) adrenergic; A1 adenosine (A(1)-R); 5-hydroxytryptamine(1A) (5-HT(1A)-R); mu opioid. Membranes were reconstituted with Galpha(i)(1) and five gamma subtypes (dimerized to beta1). Using a sensitive alpha-betagamma binding assay, we show that all recombinant betagamma (except beta1gamma1) had comparable affinity for alpha(i)(1). Using high affinity agonist binding as a measure of receptor-G protein coupling, betagamma-containing gamma11 was the most potent for A(1)-R and 5-HT(1A)-R (p < 0.05, one way ANOVA) while gamma7 was most potent for the other two receptors. gamma11 was 3-8-fold more potent for the A(1)-R than were the other gamma subtypes. Also, gamma11 was 2-8-fold more potent for A(1)-R than at the other receptors, suggesting a unique coupling specificity of the A(1)-R for gamma11. In contrast, the discrimination by receptors for the other betagamma subtypes (beta1 and gamma1, gamma2, gamma7, and gamma10) was limited (2-3-fold). Thus the exquisite betagamma specificity of individual receptors reported in whole cell studies may depend on in vivo mechanisms beyond direct receptor recognition of betagamma subtypes.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, Purinergic P1/metabolism , Receptors, Serotonin/metabolism , Animals , CHO Cells , Cell Line , Cricetinae , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Opioid, mu/metabolism , Receptors, Serotonin, 5-HT1 , Swine , Tumor Cells, Cultured
15.
Mol Cell ; 7(3): 651-60, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11463389

ABSTRACT

Agonist-evoked [Ca2+]i oscillations have been considered a biophysical phenomenon reflecting the regulation of the IP3 receptor by [Ca2+]i. Here we show that [Ca2+]i oscillations are a biochemical phenomenon emanating from regulation of Ca2+ signaling by the regulators of G protein signaling (RGS) proteins. [Ca2+]i oscillations evoked by G protein-coupled receptors require the action of RGS proteins. Inhibition of endogenous RGS protein action disrupted agonist-evoked [Ca2+]i oscillations by a stepwise conversion to a sustained response. Based on these findings and the effect of mutant RGS proteins and anti-RGS protein antibodies on Ca2+ signaling, we propose that RGS proteins within the G protein-coupled receptor complexes provide a biochemical control of [Ca2+]i oscillations.


Subject(s)
Calcium Signaling , Calcium/metabolism , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/metabolism , RGS Proteins/metabolism , Amino Acid Substitution/genetics , Animals , Calcium Signaling/drug effects , Carbachol/pharmacology , Cells, Cultured , Cholecystokinin/pharmacology , Electrophysiology , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , Macromolecular Substances , Models, Biological , Mutation/genetics , Pancreas/cytology , Pancreas/drug effects , Pancreas/metabolism , RGS Proteins/antagonists & inhibitors , RGS Proteins/genetics , Rats
16.
Fundam Clin Pharmacol ; 15(6): 393-400, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11860527

ABSTRACT

The modulation of dopamine release from the guinea-pig retina was studied using maximally effective concentrations of 10 agonists acting on G(i)-, G(s)- or G(q)-protein-coupled receptors (PCRs). Retinal discs were preincubated with [(3)H]noradrenaline and superfused; tritium overflow was evoked electrically. The following compounds acting on G(i)-PCRs reduced the tritium overflow, which represents quasi-physiological dopamine release under the experimental conditions of our study: the dopamine and alpha(2)-adrenoceptor agonist B-HT 920 by 95%, the muscarinic agonist oxotremorine by 96%, melatonin by 94%, the cannabinoid agonist WIN 55,212-2 by 71% and histamine by 66%. Tritium overflow was not affected by serotonin or by agonists acting on G(s)-PCRs (ACTH1-24 and the beta-adrenoceptor agonist procaterol) and G(q)-PCRs (angiotensin II and bradykinin). The effects of B-HT 920, oxotremorine and melatonin were studied in more detail using appropriate antagonists. The inhibitory effect of a submaximally active concentration of B-HT 920 was counteracted by the dopamine D(2/3) antagonist haloperidol but not affected by the alpha(2)-adrenoceptor antagonist phentolamine. The muscarinic antagonist atropine shifted to the right the concentration-response curve of oxotremorine (pA(2) 8.7) and the melatonin MT(2) antagonist 4-P-PDOT produced a rightward shift of the concentration-response curve of melatonin (pA(2) 10.6). Melatonin was also studied in superfused brain slices (from the guinea-pig) preincubated with [(3)H]noradrenaline. The electrically evoked tritium overflow in cerebrocortical, hippocampal and hypothalamic slices (representing quasi-physiological noradrenaline release) and in striatal slices (representing quasi-physiological dopamine release) was not affected by melatonin at a concentration that causes the maximum effect in retinal discs. In conclusion, dopamine release in the guinea-pig retina is inhibited via G(i)-PCRs including dopamine (D(2/3)), muscarinic and melatonin (MT(2)) receptors but not affected via any of the G(s)- or G(q)-PCRs under study. Unlike in the retina, melatonin fails to inhibit monoamine release in four brain regions of the guinea-pig.


Subject(s)
Dopamine/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Protein alpha Subunits, Gs/physiology , Heterotrimeric GTP-Binding Proteins/physiology , Receptors, Presynaptic/physiology , Retina/metabolism , Animals , Brain/drug effects , Brain/metabolism , Electric Stimulation , GTP-Binding Protein alpha Subunits, Gi-Go/agonists , GTP-Binding Protein alpha Subunits, Gq-G11 , GTP-Binding Protein alpha Subunits, Gs/agonists , Guinea Pigs , Heterotrimeric GTP-Binding Proteins/agonists , In Vitro Techniques , Male , Receptors, Presynaptic/agonists , Retina/drug effects
17.
J Bone Miner Res ; 15(9): 1697-706, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10976990

ABSTRACT

The mechanisms by which Gi and Gq protein- coupled receptors mediate mitogenic signaling in osteoblast-like cells are unknown and were investigated in MC3T3-E1 cells using specific receptor agonists such as lysophosphatidic acid (LPA) and prostaglandin F2alpha (PGF2alpha). In contrast to their implication in epidermal growth factor (EGF) receptor tyrosine kinase signaling, the adaptor protein Shc, the Grb2/Sos complex, and the small G protein Ras were not involved in the activation of Erk induced by either LPA or PGF2alpha in MC3T3-E1 cells, suggesting that activation of Erk by Gi and Gq protein-coupled receptors is Ras independent in these cells. Using specific kinase inhibitors and kinetic analyses, we provide evidence for two distinct components in the activation of Erk by Gi and Gq protein-coupled receptors in MC3T3-E1 cells including an Src-like kinase-dependent pathway and a protein kinase C (PKC)-dependent mechanism. Functional analyses suggested that these two components are required for optimal DNA synthesis in response to LPA and PGF2alpha. These results suggest the implication of two pathways in the stimulation of Erk and cell replication by growth factors acting through Gi and Gq protein-coupled receptors in bone-forming cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Heterotrimeric GTP-Binding Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Cell Division/drug effects , Cell Line , DNA/biosynthesis , Dinoprost/pharmacology , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , GRB2 Adaptor Protein , GTP-Binding Protein alpha Subunits, Gi-Go/agonists , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heterotrimeric GTP-Binding Proteins/agonists , Lysophospholipids/pharmacology , Mice , Osteoblasts/drug effects , Osteoblasts/enzymology , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, Cell Surface/agonists , Shc Signaling Adaptor Proteins , Signal Transduction/drug effects , Src Homology 2 Domain-Containing, Transforming Protein 1 , Virulence Factors, Bordetella/pharmacology
18.
Biochem Biophys Res Commun ; 272(3): 922-8, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10860852

ABSTRACT

We have characterized vasoactive intestinal peptide (VIP) receptor/G-protein coupling in rat alveolar macrophage (AM) membranes and find that pertussis toxin treatment and antisera against G(alphai3) and G(alphas) reduce high-affinity (125)I-VIP binding, indicating that both G(alphas) and G(alphai3) couple to the VIP-receptor. The predominant VIP-receptor subtype in AM is VPAC(1) and we examined the G-protein interactions of the human VPAC(1) that had been transfected into HEK293 cells. VPAC(1) has a molecular mass of 56 kDa; GTP analogs reduced (125)I-VIP binding to this protein demonstrating that high-affinity binding of VIP to the receptor requires coupling to G-protein. Functional VIP/VPAC(1)/G-protein complexes were captured by covalent cross-linking and analyzed by Western blotting. The transfected human VPAC(1) receptor in HEK293 was found to be coupled to G(alphas) but not G(alphai) or G(alphaq). Furthermore, pertussis toxin treatment had no effect on VPAC(1)/G-protein coupling in these cells. These observations suggest that the G-proteins activated by VPAC(1) may be dependent upon species and cell type.


Subject(s)
Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/metabolism , Macrophages, Alveolar/drug effects , Receptors, Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/pharmacology , Animals , Binding Sites/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cross-Linking Reagents/metabolism , Enzyme Activation/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/agonists , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/agonists , GTP-Binding Protein alpha Subunits, Gs/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gs/metabolism , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/pharmacology , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Humans , Immune Sera/pharmacology , Macrophages, Alveolar/cytology , Macrophages, Alveolar/metabolism , Male , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Organ Specificity , Pertussis Toxin , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Vasoactive Intestinal Peptide/chemistry , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I , Signal Transduction/drug effects , Species Specificity , Virulence Factors, Bordetella/pharmacology
19.
Biochem Biophys Res Commun ; 272(3): 959-65, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10860858

ABSTRACT

The binding of [(125)I] orexin-A (Ox-A) to particulates from Chinese hamster ovary (CHO) cells expressing the cloned orexin-A receptor, or from rat forebrain areas, was sensitive to blockers of phosphatidylinositol-specific phospholipase C (PtdIns-PLC) U-73122 and ET-18-OCH(3), little affected by phospholipase A(2) inhibitor quinacrine, and not sensitive to D609, a xanthate inhibitor of phosphatidylcholine-selective PLC. Interaction of the receptor with a PtdIns-PLC was further indicated by a large sensitivity of the binding to Ca(2+). Up to 50% of the binding was sensitive to the G-protein nucleotide site agonist GTP-gamma-S. Ligand attachment to the orexin-A receptor thus depends on an association with both PtdIns-PLC and G-protein alpha-subunits. In all paradigms examined, the binding of [(125)I]orexin-A was competed by human/rat neuropeptide Y (hNPY) and porcine secretin with a potency similar to orexin-A (IC(50) range 30-100 nM). The rank order of potency for NPY-related peptides was hNPY > porcine peptide YY (pPYY) > (Leu(31), Pro(34)) human PYY > human PYY(3-36) > hNPY free acid > human pancreatic polypeptide. Among secretin-related peptides, the rank order of potency was porcine secretin > or = orexin-A > human pituitary adenylate cyclase-activating peptide > orexin-B > porcine vasoactive intestinal peptide. Among opioid peptides, rat beta-endorphin and camel delta-endorphin were much less active than NPY and secretin, and two enkephalins were inactive at 1 microM. In view of high abundance of NPY in forebrain, the above cross-reactivity could indicate a significant contribution of NPY to signaling via orexin-A receptors.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Enzyme Inhibitors/metabolism , Intracellular Signaling Peptides and Proteins , Neuropeptide Y/metabolism , Neuropeptides/antagonists & inhibitors , Neuropeptides/metabolism , Secretin/metabolism , Type C Phospholipases/antagonists & inhibitors , Animals , Binding, Competitive , CHO Cells , Calcium/metabolism , Calcium/pharmacology , Cricetinae , Enzyme Inhibitors/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Inhibitory Concentration 50 , Magnesium/metabolism , Magnesium/pharmacology , Male , Orexin Receptors , Orexins , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Phospholipases A/antagonists & inhibitors , Prosencephalon/drug effects , Prosencephalon/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Signal Transduction/drug effects , Type C Phospholipases/metabolism
20.
Leuk Lymphoma ; 38(1-2): 39-48, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10811446

ABSTRACT

We have reviewed the current knowledge on CXC chemokine interleukin-8 (IL-8) and human hematopoiesis, and more generally on agonists of heterotrimeric Gi2 proteins as regulators of human hematopoiesis. It appears that low doses of IL-8, a Gi2-agonist produced in an autocrine fashion by normal hematopoietic progenitors, mature blood cells and leukemic cells, promotes cell survival or/and proliferation in response to hematopoietic cytokines. More importantly, inactivation of the IL-8/Gi2 pathways inhibits CD34+ cell proliferation and colony formation. Similar positive effects on hematopoiesis of other, physiological or pathological, agonists of Gi2 proteins are discussed, as well as the molecular pathways involved and the consequences of activation of other G proteins (Gq, G16) by IL-8 and other Gi2-agonists.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Heterotrimeric GTP-Binding Proteins/physiology , Interleukin-8/physiology , Macrophage Colony-Stimulating Factor/physiology , Proto-Oncogene Proteins/physiology , Autocrine Communication , Drug Synergism , GTP-Binding Protein alpha Subunit, Gi2 , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Heterotrimeric GTP-Binding Proteins/agonists , Humans , Interleukin-8/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Paracrine Communication , Proto-Oncogene Proteins/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...