Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Complementary Medicines
Therapeutic Methods and Therapies TCIM
Country/Region as subject
Publication year range
1.
J Med Chem ; 64(9): 5905-5930, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33904304

ABSTRACT

There is an urgent need for new treatments for visceral leishmaniasis (VL), a parasitic infection which impacts heavily large areas of East Africa, Asia, and South America. We previously reported on the discovery of GSK3494245/DDD01305143 (1) as a preclinical candidate for VL and, herein, we report on the medicinal chemistry program that led to its identification. A hit from a phenotypic screen was optimized to give a compound with in vivo efficacy, which was hampered by poor solubility and genotoxicity. The work on the original scaffold failed to lead to developable compounds, so an extensive scaffold-hopping exercise involving medicinal chemistry design, in silico profiling, and subsequent synthesis was utilized, leading to the preclinical candidate. The compound was shown to act via proteasome inhibition, and we report on the modeling of different scaffolds into a cryo-EM structure and the impact this has on our understanding of the series' structure-activity relationships.


Subject(s)
Drug Design , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemistry , Protozoan Proteins/metabolism , Animals , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/metabolism , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Binding Sites , Cell Line , Drug Evaluation, Preclinical , Half-Life , Humans , Leishmania donovani/drug effects , Leishmania donovani/metabolism , Leishmaniasis, Visceral/drug therapy , Leishmaniasis, Visceral/parasitology , Mice , Molecular Dynamics Simulation , Proteasome Endopeptidase Complex/chemistry , Proteasome Inhibitors/metabolism , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Protein Subunits/chemistry , Protein Subunits/metabolism , Protozoan Proteins/chemistry , Pyridines/chemistry , Pyridines/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Solubility , Structure-Activity Relationship
2.
Nature ; 584(7820): 304-309, 2020 08.
Article in English | MEDLINE | ID: mdl-32581365

ABSTRACT

The human GABAB receptor-a member of the class C family of G-protein-coupled receptors (GPCRs)-mediates inhibitory neurotransmission and has been implicated in epilepsy, pain and addiction1. A unique GPCR that is known to require heterodimerization for function2-6, the GABAB receptor has two subunits, GABAB1 and GABAB2, that are structurally homologous but perform distinct and complementary functions. GABAB1 recognizes orthosteric ligands7,8, while GABAB2 couples with G proteins9-14. Each subunit is characterized by an extracellular Venus flytrap (VFT) module, a descending peptide linker, a seven-helix transmembrane domain and a cytoplasmic tail15. Although the VFT heterodimer structure has been resolved16, the structure of the full-length receptor and its transmembrane signalling mechanism remain unknown. Here we present a near full-length structure of the GABAB receptor, captured in an inactive state by cryo-electron microscopy. Our structure reveals several ligands that preassociate with the receptor, including two large endogenous phospholipids that are embedded within the transmembrane domains to maintain receptor integrity and modulate receptor function. We also identify a previously unknown heterodimer interface between transmembrane helices 3 and 5 of both subunits, which serves as a signature of the inactive conformation. A unique 'intersubunit latch' within this transmembrane interface maintains the inactive state, and its disruption leads to constitutive receptor activity.


Subject(s)
Cryoelectron Microscopy , Receptors, GABA-B/chemistry , Receptors, GABA-B/ultrastructure , Calcium/metabolism , Ethanolamines/chemistry , Ethanolamines/metabolism , Humans , Ligands , Models, Molecular , Phosphorylcholine/chemistry , Phosphorylcholine/metabolism , Protein Domains , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Receptors, GABA-B/metabolism , Structure-Activity Relationship
3.
Plant Cell ; 31(12): 2855-2867, 2019 12.
Article in English | MEDLINE | ID: mdl-31628168

ABSTRACT

Actins are among the most abundant and conserved proteins in eukaryotic cells, where they form filamentous structures that perform vital roles in key cellular processes. Although large amounts of data on the biochemical activities, dynamic behaviors, and important cellular functions of plant actin filaments have accumulated, their structural basis remains elusive. Here, we report a 3.9 Å structure of the plant actin filament from Zea mays pollen (ZMPA) using cryo-electron microscopy. The structure shows a right-handed, double-stranded (two parallel strands) and staggered architecture that is stabilized by intra- and interstrand interactions. While the overall structure resembles that of other actin filaments, its DNase I binding loop bends farther outward, adopting an open conformation similar to that of the jasplakinolide- or beryllium fluoride (BeFx)-stabilized rabbit skeletal muscle actin (RSMA) filament. Single-molecule magnetic tweezers analysis revealed that the ZMPA filament can resist a greater stretching force than the RSMA filament. Overall, these data provide evidence that plant actin filaments have greater stability than animal actin filaments, which might be important to their role as tracks for long-distance vesicle and organelle transportation.plantcell;31/12/2855/FX1F1fx1.


Subject(s)
Actin Cytoskeleton/chemistry , Pollen/chemistry , Zea mays/chemistry , Actin Cytoskeleton/metabolism , Cryoelectron Microscopy , Hydrophobic and Hydrophilic Interactions , Pollen/metabolism , Protein Conformation , Protein Subunits/chemistry , Zea mays/metabolism
4.
Structure ; 27(7): 1171-1183.e3, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31130483

ABSTRACT

Nicotinic acetylcholine receptors (nAChRs) modulate synaptic transmission in the nervous system. These receptors have emerged as therapeutic targets in drug discovery for treating several conditions, including Alzheimer's disease, pain, and nicotine addiction. In this in silico study, we use a combination of equilibrium and nonequilibrium molecular dynamics simulations to map dynamic and structural changes induced by nicotine in the human α4ß2 nAChR. They reveal a striking pattern of communication between the extracellular binding pockets and the transmembrane domains (TMDs) and show the sequence of conformational changes associated with the initial steps in this process. We propose a general mechanism for signal transduction for Cys-loop receptors: the mechanistic steps for communication proceed firstly through loop C in the principal subunit, and are subsequently transmitted, gradually and cumulatively, to loop F of the complementary subunit, and then to the TMDs through the M2-M3 linker.


Subject(s)
Lipid Bilayers/chemistry , Nicotine/chemistry , Phosphatidylcholines/chemistry , Protein Subunits/chemistry , Receptors, Nicotinic/chemistry , Signal Transduction , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Humans , Hydrogen-Ion Concentration , Ligands , Lipid Bilayers/metabolism , Molecular Dynamics Simulation , Nicotine/metabolism , Phosphatidylcholines/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Subunits/metabolism , Receptors, Nicotinic/metabolism , Thermodynamics
5.
Nucleic Acids Res ; 47(8): 3904-3920, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30793213

ABSTRACT

Understanding of mechanistic details of Mediator functioning in plants is impeded as the knowledge of subunit organization and structure is lacking. In this study, an interaction map of Arabidopsis Mediator complex was analyzed to understand the arrangement of the subunits in the core part of the complex. Combining this interaction map with homology-based modeling, probable structural topology of core part of the Arabidopsis Mediator complex was deduced. Though the overall topology of the complex was similar to that of yeast, several differences were observed. Many interactions discovered in this study are not yet reported in other systems. AtMed14 and AtMed17 emerged as the key component providing important scaffold for the whole complex. AtMed6 and AtMed10 were found to be important for linking head with middle and middle with tail, respectively. Some Mediator subunits were found to form homodimers and some were found to possess transactivation property. Subcellular localization suggested that many of the Mediator subunits might have functions beyond the process of transcription. Overall, this study reveals role of individual subunits in the organization of the core complex, which can be an important resource for understanding the molecular mechanism of functioning of Mediator complex and its subunits in plants.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis/genetics , Gene Expression Regulation, Plant , Mediator Complex/chemistry , Protein Interaction Mapping , Protein Subunits/chemistry , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Binding Sites , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Mediator Complex/genetics , Mediator Complex/metabolism , Models, Molecular , Onions/genetics , Onions/metabolism , Plant Cells/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Seedlings/genetics , Seedlings/metabolism , Structural Homology, Protein
6.
Comput Biol Chem ; 72: 33-44, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29346071

ABSTRACT

Identification of hit compounds against specific target form the starting point for a drug discovery program. A consistent decline of new chemical entities (NCEs) in recent years prompted a challenge to explore newer approaches to discover potential hit compounds that in turn can be converted into leads, and ultimately drug with desired therapeutic efficacy. The vast amount of omics and activity data available in public databases offers an opportunity to identify novel targets and their potential inhibitors. State of the art in silico methods viz., clustering of compounds, virtual screening, molecular docking, MD simulations and MMPBSA calculations were employed in a pipeline to identify potential 'hits' against those targets as well whose structures, as of now, could only predict through threading approaches. In the present work, we have started from scratch, amino acid sequence of target and compounds retrieved from PubChem compound database, modeled it in such a way that led to the identification of possible inhibitors of Dam1 complex subunit Ask1 of Candida albicans. We also propose a ligand based binding site determination approach. We have identified potential inhibitors of Ask1 subunit of a Dam1 complex of C. albicans, which is required to prevent precocious spindle elongation in pre-mitotic phases. The proposed scheme may aid to find virtually potential inhibitors of other unique targets against candida.


Subject(s)
Antifungal Agents/chemistry , Fungal Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/antagonists & inhibitors , Protein Subunits/antagonists & inhibitors , Small Molecule Libraries/chemistry , Candida albicans , Drug Evaluation, Preclinical , Fungal Proteins/chemistry , Hydrogen Bonding , Ligands , Microtubule-Associated Proteins/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Protein Subunits/chemistry
7.
Eur J Nutr ; 57(3): 1157-1168, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28324208

ABSTRACT

PURPOSE: This study examined the effect of soy proteins with depletion of different subunits of the two major storage proteins, ß-conglycinin and glycinin, on hepatic lipids and proteins involved in lipid metabolism in rats, since the bioactive component of soy responsible for lipid-lowering is unclear. METHODS: Weanling Sprague Dawley rats were fed diets containing either 20% casein protein in the absence (casein) or presence (casein + ISF) of isoflavones or 20% alcohol-washed soy protein isolate (SPI) or 20% soy protein concentrates derived from a conventional (Haro) or 2 soybean lines lacking the α' subunit of ß-conglycinin and the A1-3 (1TF) or A1-5 (1a) subunits of glycinin. After 8 weeks, the rats were necropsied and liver proteins and lipids were extracted and analysed. RESULTS: The results showed that soy protein diets reduced lipid droplet accumulation and content in the liver compared to casein diets. The soy protein diets also decreased the level of hepatic mature SREBP-1 and FAS in males, with significant decreases in diets 1TF and 1a compared to the casein diets. The effect of the soy protein diets on female hepatic mature SREBP-1, FAS, and HMGCR was confounded since casein + ISF decreased these levels compared to casein alone perhaps muting the decrease by soy protein. A reduction in both phosphorylated and total STAT3 in female livers by ISF may account for the gender difference in mechanism in the regulation and protein expression of the lipid modulators. CONCLUSIONS: Overall, soy protein deficient in the α' subunit of ß-conglycinin and A1-5 subunits of glycinin maintain similar hypolipidemic function compared to the conventional soy protein. The exact bioactive component(s) warrant identification.


Subject(s)
Antigens, Plant/therapeutic use , Globulins/therapeutic use , Hyperlipidemias/prevention & control , Lipid Metabolism , Liver/metabolism , Plant Proteins, Dietary/therapeutic use , Protein Subunits/therapeutic use , Seed Storage Proteins/therapeutic use , Soybean Proteins/therapeutic use , Animals , Antigens, Plant/chemistry , Antigens, Plant/genetics , Antigens, Plant/metabolism , Caseins/adverse effects , Diet, High-Fat/adverse effects , Female , Food, Genetically Modified , Globulins/chemistry , Globulins/genetics , Globulins/metabolism , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Lipid Droplets/metabolism , Lipid Droplets/pathology , Liver/enzymology , Liver/pathology , Male , Phosphorylation , Plant Proteins, Dietary/chemistry , Plant Proteins, Dietary/genetics , Plant Proteins, Dietary/metabolism , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Protein Processing, Post-Translational , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Seed Storage Proteins/chemistry , Seed Storage Proteins/genetics , Seed Storage Proteins/metabolism , Sex Characteristics , Soybean Proteins/chemistry , Soybean Proteins/genetics , Soybean Proteins/metabolism , Vacuoles/pathology , Weaning
8.
Acc Chem Res ; 50(2): 331-340, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28177216

ABSTRACT

Despite its essentiality to life, iron presents significant challenges to cells: the exceedingly low solubility of Fe3+ limits its bioavailability, and the reactivity of Fe2+ toward H2O2 is a source of the toxic hydroxyl radical (HO•). Consequently, cellular levels of free iron are highly regulated to ensure sufficiency while preventing iron-induced toxicity. Relatively little is known about the fate of iron in the bacterial cytosol or how cells balance the need for relatively high cytosolic iron concentrations with the potential toxicity of the nutrient. Iron storage proteins are integral to iron metabolism, and bacteria utilize two types of ferritin-like molecules to store iron, bacterial ferritin (Ftn) and bacterioferritin (Bfr). Ftn and Bfr compartmentalize iron at concentrations far above the solubility of Fe3+ and protect the reducing cell environment from unwanted Fe3+/Fe2+ redox cycling. This Account focuses on our laboratory's efforts to study iron storage proteins in the model bacterium Pseudomonas aeruginosa, an opportunistic pathogen. Prior to our studies, it was thought that P. aeruginosa cells relied on a single Bfr assembled from two distinct subunits coded by the bfrA and bfrB genes. It is now known that, like in most bacteria, two iron storage proteins coexist in P. aeruginosa cells, a bacterial Ftn (FtnA), coded by the ftnA (formerly bfrA) gene and a bacterioferritin (BfrB), coded by the bfrB gene. Studies with BfrB showed that Fe2+ oxidation occurs at ferroxidase centers (FCs), followed by gated translocation of Fe3+ to the interior cavity, a process that is, surprisingly, distinct from that observed with the extensively studied Bfr from Escherichia coli, where the FCs are stable and function only as a catalytic site for O2 reduction. Investigations with BfrB showed that the oxidation of Fe2+ at FCs and the internalization of Fe3+ depend on long-range cooperative motions, extending from 4-fold pores, via B-pores, into FCs. It remains to be seen whether similar studies with E. coli Bfr will reveal distinct cooperative motions contributing to the stability of its FCs. Mobilization of Fe3+ stored in BfrB requires interaction with a ferredoxin (Bfd), which transfers electrons to reduce Fe3+ in the internal cavity of BfrB for subsequent release of Fe2+. The structure of the BfrB/Bfd complex furnished the only known structure of a ferritin molecule in complex with a physiological protein partner. The BfrB/Bfd complex is stabilized by hot-spot residues in both proteins, which interweave into a highly complementary hot region. The hot-spot residues are conserved in the sequences of Bfr and Bfd proteins from a number of bacteria, indicating that the BfrB/Bfd interaction is of widespread significance in bacterial iron metabolism. The BfrB/Bfd structure also furnished the only known structure of a Bfd, which revealed a novel helix-turn-helix fold different from the ß-strand and α-helix fold of plant and vertebrate [2Fe-2S]-ferredoxins. Bfds seem to be unique to bacteria; consequently, although mobilization of iron from eukaryotic ferritins may also be facilitated by protein-protein interactions, the nature of the protein that delivers electrons to the ferric core of eukaryotic ferritins remains unknown.


Subject(s)
Bacterial Proteins/chemistry , Cytochrome b Group/chemistry , Ferritins/chemistry , Iron/chemistry , Bacterial Proteins/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Cytochrome b Group/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Ferritins/metabolism , Iron/metabolism , Molecular Dynamics Simulation , Oxidation-Reduction , Protein Interaction Domains and Motifs , Protein Subunits/chemistry , Protein Subunits/metabolism , Pseudomonas aeruginosa/metabolism
9.
Acta Pharmacol Sin ; 37(7): 908-18, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27238210

ABSTRACT

AIM: Bufalin is one of the active components in the traditional Chinese medicine ChanSu that is used to treat arrhythmia, inflammation and cancer. BF211 is a bufalin derivative with stronger cytotoxic activity in cancer cells. The aim of this study was to identify the putative target proteins of BF211 and the signaling pathways in cancer cells. METHODS: A549 human lung cancer cells were treated with BF211. A SILAC-based proteomic analysis was used to detect the protein expression profiles of BF211-treated A549 cells. Cellular proteasome activities were examined using fluorogenic peptide substrates, and the binding affinities of BF211 to recombinant proteasome subunit proteins were evaluated using the Biacore assay. The expression levels of proteasome subunits were determined using RT-PCR and Western blotting, and the levels of the integral 26S proteasome were evaluated using native PAGE analysis. RESULTS: The proteomic analysis revealed that 1282 proteins were differentially expressed in BF211-treated A549 cells, and the putative target proteins of BF211 were associated with various cellular functions, including transcription, translation, mRNA splicing, ribosomal protein synthesis and proteasome function. In A549 cells, BF211 (5, 10, and 20 nmol/L) dose-dependently inhibited the enzymatic activities of proteasome. But BF211 displayed a moderate affinity in binding to proteasome ß1 subunit and no binding affinity to the ß2 and ß5 subunits. Moreover, BF211 (0.1, 1, and 10 nmol/L) did not inhibit the proteasome activities in the cell lysates. BF211 (5, 10, and 20 nmol/L) significantly decreased the expression level of proteasome ß1 subunit and the levels of integral 26S proteasome in A549 cells. Similarly, knockdown of the ß1 subunit with siRNA in A549 cells significantly decreased integral 26S proteasome and proteasome activity. CONCLUSION: BF211 inhibits proteasome activity in A549 cells by decreasing ß1 subunit expression and disrupting proteasome assembly.


Subject(s)
Bufanolides/pharmacology , Lung Neoplasms/enzymology , Piperazines/pharmacology , Proteasome Endopeptidase Complex/biosynthesis , Proteasome Endopeptidase Complex/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Lung Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , Proteomics , RNA, Small Interfering/pharmacology
10.
J Physiol ; 594(19): 5555-71, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27195487

ABSTRACT

KEY POINTS: The role of the ß1 strand in GABAA receptor function is unclear. It lies anti-parallel to the ß2 strand, which is known to participate in receptor activation. Molecular dynamics simulation revealed solvent accessible residues within the ß1 strand of the GABAA ß3 homopentamer that might be amenable to analysis using the substituted Cys accessibility method. Cys substitutions from Asp43 to Thr47 in the GABAA α1 subunit showed that D43C and T47C reduced the apparent potency of GABA. F45C caused a biphasic GABA concentration-response relationship and increased spontaneous gating. Cys43 and Cys47 were accessible to 2-aminoethyl methanethiosulphonate (MTSEA) modification, whereas Cys45 was not. Both GABA and the allosteric agonist propofol reduced MTSEA modification of Cys43 and Cys47. By contrast, modification of Cys64 in the ß2 strand loop D was impeded by GABA but unaffected by propofol. These data reveal movement of ß1 strand loop G residues during agonist activation of the GABAA receptor. ABSTRACT: The GABAA receptor α subunit ß1 strand runs anti-parallel to the ß2 strand, which contains loop D, known to participate in receptor activation and agonist binding. However, a role for the ß1 strand has yet to be established. We used molecular dynamics simulation to quantify the solvent accessible surface area (SASA) of ß1 strand residues in the GABAA ß3 homopentamer structure. Residues in the complementary interface equivalent to those between Asp43 and Thr47 in the α1 subunit have an alternating pattern of high and low SASA consistent with a ß strand structure. We investigated the functional role of these ß1 strand residues in the α1 subunit by individually replacing them with Cys residues. D43C and T47C substitutions reduced the apparent potency of GABA at α1ß2γ2 receptors by 50-fold and eight-fold, respectively, whereas the F45C substitution caused a biphasic GABA concentration-response relationship and increased spontaneous gating. Receptors with D43C or T47C substitutions were sensitive to 2-aminoethyl methanethiosulphonate (MTSEA) modification. However, GABA-evoked currents mediated by α1(F45C)ß2γ2 receptors were unaffected by MTSEA, suggesting that this residue is inaccessible. Both GABA and the allosteric agonist propofol reduced MTSEA modification of α1(D43C)ß2γ2 and α1(T47C)ß2γ2 receptors, indicating movement of the ß1 strand even during allosteric activation. This is in contrast to α1(F64C)ß2γ2 receptors, where only GABA, but not propofol, reduced MTSEA modification. These findings provide the first functional evidence for movement of the ß1 strand during gating of the receptor and identify residues that are critical for maintaining GABAA receptor function.


Subject(s)
Receptors, GABA-A/chemistry , Receptors, GABA-A/physiology , Ethyl Methanesulfonate/analogs & derivatives , Ethyl Methanesulfonate/pharmacology , HEK293 Cells , Humans , Molecular Dynamics Simulation , Propofol/pharmacology , Protein Conformation, beta-Strand , Protein Subunits/chemistry , Protein Subunits/physiology , gamma-Aminobutyric Acid/pharmacology
11.
Nature ; 525(7567): 68-72, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26280334

ABSTRACT

Phosphorus is required for all life and microorganisms can extract it from their environment through several metabolic pathways. When phosphate is in limited supply, some bacteria are able to use phosphonate compounds, which require specialized enzymatic machinery to break the stable carbon-phosphorus (C-P) bond. Despite its importance, the details of how this machinery catabolizes phosphonates remain unknown. Here we determine the crystal structure of the 240-kilodalton Escherichia coli C-P lyase core complex (PhnG-PhnH-PhnI-PhnJ; PhnGHIJ), and show that it is a two-fold symmetric hetero-octamer comprising an intertwined network of subunits with unexpected self-homologies. It contains two potential active sites that probably couple phosphonate compounds to ATP and subsequently hydrolyse the C-P bond. We map the binding site of PhnK on the complex using electron microscopy, and show that it binds to a conserved insertion domain of PhnJ. Our results provide a structural basis for understanding microbial phosphonate breakdown.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Lyases/chemistry , Lyases/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Biocatalysis , Carbon/chemistry , Carbon/metabolism , Conserved Sequence , Crystallography, X-Ray , Escherichia coli Proteins/ultrastructure , Hydrolysis , Iron/chemistry , Iron/metabolism , Lyases/ultrastructure , Microscopy, Electron , Models, Molecular , Organophosphonates/metabolism , Phosphorus/chemistry , Phosphorus/metabolism , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , Sulfur/chemistry , Sulfur/metabolism
12.
Mol Immunol ; 67(2 Pt B): 642-51, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26319314

ABSTRACT

Ferritin, an evolutionarily conserved iron-binding protein, plays important roles in iron storage and detoxification and in host immune response to invading stimulus as well. In the present study, we identified three ferritin subunit analog cDNAs from Chinese giant salamander (Andrias davidianus). All the three ferritin subunit cDNAs had a putative iron responsive element in the 5'-untranslated region. Two deduced ferritin subunits (designated as cgsFerH and cgsFerM) had the highest identity of 90% to H type subunit of vertebrate ferritins, while another deduced ferritin subunit (designated as cgsFerL) had the highest identity of 84% to L type subunit of vertebrate ferritins. The Chinese giant salamander ferritin (cgsFer) was widely expressed in various tissues, with highest expression for cgsFerH and cgsFerL in liver and highest expression for cgsFerM in spleen. Infection of Chinese giant salamander with A. davidianus ranavirus showed significant induction of cgsFer expression. Both lipopolysaccharide and iron challenge drastically augmented cgsFer expression in the splenocytes and hepatocytes from Chinese giant salamander. In addition, recombinant cgsFers bound to ferrous iron in a dose-dependent manner, with significant ferroxidase activity. Furthermore, the recombinant cgsFer inhibited the growth of the pathogen Vibrio anguillarum. These results indicated that cgsFer was potential candidate of immune molecules involved in acute phase response to invading microbial pathogens in Chinese giant salamander possibly through its regulatory roles in iron homeostasis.


Subject(s)
Ferritins/metabolism , Urodela/metabolism , Urodela/microbiology , Vibrio/physiology , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary/genetics , Electrophoresis, Polyacrylamide Gel , Ferritins/chemistry , Ferritins/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Iron/pharmacology , Jurkat Cells , Lipopolysaccharides/pharmacology , Molecular Sequence Data , Phylogeny , Protein Subunits/analogs & derivatives , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Response Elements/genetics , Sequence Alignment , Sequence Analysis, Protein , Tissue Distribution/drug effects , Urodela/genetics , Vibrio/drug effects
13.
Rev. bras. anestesiol ; 65(3): 207-212, May-Jun/2015. tab, graf
Article in English | LILACS | ID: lil-748922

ABSTRACT

BACKGROUND AND OBJECTIVE: Postoperative pain treatment in mastectomy remains a major challenge despite the multimodal approach. The aim of this study was to investigate the analgesic effect of intravenous lidocaine in patients undergoing mastectomy, as well as the postoperative consumption of opioids. METHODS: After approval by the Human Research Ethics Committee of the Instituto de Medicina Integral Prof. Fernando Figueira in Recife, Pernambuco, a randomized, blind, controlled trial was conducted with intravenous lidocaine at a dose of 3 mg/kg infused over 1 h in 45 women undergoing mastectomy under general anesthesia. One patient from placebo group was. RESULTS: Groups were similar in age, body mass index, type of surgery, and postoperative need for opioids. Two of 22 patients in lidocaine group and three of 22 patients in placebo group requested opioid (p = 0.50). Pain on awakening was identified in 4/22 of lidocaine group and 5/22 of placebo group (p = 0.50); in the post-anesthetic recovery room in 14/22 and 12/22 (p = 0.37) of lidocaine and placebo groups, respectively. Pain evaluation 24 h after surgery showed that 2/22 and 3/22 patients (p = 0.50) of lidocaine and placebo groups, respectively, complained of pain. CONCLUSION: Intravenous lidocaine at a dose of 3 mg/kg administered over a period of an hour during mastectomy did not promote additional analgesia compared to placebo in the first 24 h, and has not decreased opioid consumption. However, a beneficial effect of intravenous lidocaine in selected and/or other therapeutic regimens patients cannot be ruled out. .


JUSTIFICATIVA E OBJETIVO: O tratamento da dor pós-operatória em mastectomia continua sendo um grande desafio apesar da abordagem multimodal. O objetivo deste estudo foi investigar o efeito analgésico da lidocaína intravenosa em pacientes submetidas a mastectomia, como também, o consumo de opioide pós-operatório. MÉTODOS: Após aprovação pelo comitê de ética e pesquisa em seres humanos do Instituto de Medicina Integral Prof. Fernando Figueira em Recife - Pernambuco foi realizado ensaio clínico aleatório encoberto placebo controlado com lidocaína intravenosa na dose de 3 mg/kg infundida em uma hora, em 45 mulheres submetidas a mastectomia sob anestesia geral. Excluída uma paciente do grupo placebo. RESULTADOS: Os grupos foram semelhantes quanto à idade, índice de massa corpórea, tipo de intervenção cirúrgica e necessidade de opioide no pós-operatório. Solicitaram opioide 2/22 pacientes nos grupos da lidocaína e 3/22 placebo (p = 0,50). Identificada a dor ao despertar em 4/22 no grupo lidocaína e 5/22 (p = 0,50) no grupo placebo; na sala de recuperação pós-anestésica em 14/22 e 12/22 (p = 0,37) nos grupos lidocaína e placebo respectivamente. Ao avaliar a dor 24 horas após o procedimento cirúrgico 3/22 e 2/22 (p = 0,50) das pacientes relataram dor em ambos os grupos respectivamente. CONCLUSÃO: A lidocaína intravenosa na dose de 3mg/kg administrada em um período de uma hora no transoperatório de mastectomia não promoveu analgesia adicional em relação ao grupo placebo nas primeiras 24 horas e não diminuiu o consumo de opioide. Contudo, um efeito benéfico da lidocaína intravenosa em pacientes selecionadas e/ou em outros regimes terapêuticos não pode ser descartado. .


JUSTIFICACIÓN Y OBJETIVO: El tratamiento del dolor postoperatorio en la mastectomía continúa siendo un gran reto a pesar del abordaje multimodal. El objetivo de este estudio fue investigar el efecto analgésico de la lidocaína intravenosa en pacientes sometidas a mastectomía, así como el consumo postoperatorio de opiáceos. MÉTODOS: Después de la aprobación por el Comité de Ética e Investigación en seres humanos del Instituto de Medicina Integral Prof. Fernando Figueira, en Recife, Pernambuco, se realizó un ensayo clínico aleatorizado, encubierto, placebo controlado con lidocaína intravenosa en una dosis de 3 mg/kg infundida en una hora, en 45 mujeres sometidas a mastectomía bajo anestesia general. Una paciente del grupo placebo fue excluida. RESULTADOS: Los grupos fueron similares en cuanto a la edad, índice de masa corporal, tipo de intervención quirúrgica y necesidad de opiáceos en el postoperatorio. Solicitaron opiáceos 2/22 pacientes en los grupos de la lidocaína y 3/22 placebo (p = 0,50). Fue identificado el dolor al despertar en 4/22 en el grupo lidocaína y 5/22 (p = 0,50) en el grupo placebo; en la sala de recuperación postanestésica en 14/22 y 12/22 (p = 0,37) en los grupos lidocaína y placebo, respectivamente. Al calcular el dolor 24 h después del procedimiento quirúrgico 3/22 y 2/22 (p = 0,50) de las pacientes relataron dolor en ambos grupos respectivamente. CONCLUSIÓN: La lidocaína intravenosa en una dosis de 3 mg/kg administrada en un período de una hora en el transoperatorio de mastectomía no generó analgesia adicional con relación al grupo placebo en las primeras 24 h y no disminuyó el consumo de opiáceos. Sin embargo, no puede ser descartado un efecto beneficioso de la lidocaína intravenosa en pacientes seleccionadas y/o en otros regímenes terapéuticos. .


Subject(s)
Humans , Metapneumovirus/genetics , Transcription, Genetic , Viral Proteins/chemistry , Amino Acid Sequence , Adenosine Monophosphate/metabolism , Crystallography, X-Ray , DNA , Edetic Acid/pharmacology , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Multimerization , Protein Stability , Protein Subunits/chemistry , RNA, Viral/metabolism , RNA, Viral/ultrastructure , Scattering, Small Angle , Solutions , Solvents , Viral Proteins/metabolism , Viral Proteins/ultrastructure , Zinc Fingers
14.
FEBS Lett ; 589(13): 1444-9, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25953126

ABSTRACT

Substrate binding properties of the large (LS) and small (SS) subunits of potato tuber ADP-glucose pyrophosphorylase were investigated by using isothermal titration calorimetry. Our results clearly show that the wild type heterotetramer (S(WT)L(WT)) possesses two distinct types of ATP binding sites, whereas the homotetrameric LS and SS variant forms only exhibited properties of one of the two binding sites. The wild type enzyme also exhibited significantly increased affinity to this substrate compared to the homotetrameric enzyme forms. No stable binding was evident for the second substrate, glucose-1-phosphate, in the presence or absence of ATPγS suggesting that interaction of glucose-1-phosphate is dependent on hydrolysis of ATP and supports the Theorell-Chance bi bi reaction mechanism.


Subject(s)
Calorimetry/methods , Glucose-1-Phosphate Adenylyltransferase/metabolism , Plant Proteins/metabolism , Plant Tubers/enzymology , Solanum tuberosum/enzymology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Binding Sites , Binding, Competitive , Blotting, Western , Glucose-1-Phosphate Adenylyltransferase/chemistry , Glucosephosphates/chemistry , Glucosephosphates/metabolism , Kinetics , Models, Molecular , Molecular Structure , Plant Proteins/chemistry , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , Substrate Specificity , Thermodynamics
15.
J Biol Chem ; 289(41): 28338-51, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25028511

ABSTRACT

Functional heterologous expression of naturally expressed mouse α6*-nicotinic acetylcholine receptors (mα6*-nAChRs; where "*" indicates the presence of additional subunits) has been difficult. Here we expressed and characterized wild-type (WT), gain-of-function, chimeric, or gain-of-function chimeric nAChR subunits, sometimes as hybrid nAChRs containing both human (h) and mouse (m) subunits, in Xenopus oocytes. Hybrid mα6mß4hß3- (∼ 5-8-fold) or WT mα6mß4mß3-nAChRs (∼ 2-fold) yielded higher function than mα6mß4-nAChRs. Function was not detected when mα6 and mß2 subunits were expressed together or in the additional presence of hß3 or mß3 subunits. However, function emerged upon expression of mα6mß2mß3(V9'S)-nAChRs containing ß3 subunits having gain-of-function V9'S (valine to serine at the 9'-position) mutations in transmembrane domain II and was further elevated 9-fold when hß3(V9'S) subunits were substituted for mß3(V9'S) subunits. Studies involving WT or gain-of-function chimeric mouse/human ß3 subunits narrowed the search for domains that influence functional expression of mα6*-nAChRs. Using hß3 subunits as templates for site-directed mutagenesis studies, substitution with mß3 subunit residues in extracellular N-terminal domain loops "C" (Glu(221) and Phe(223)), "E" (Ser(144) and Ser(148)), and "ß2-ß3" (Gln(94) and Glu(101)) increased function of mα6mß2*- (∼ 2-3-fold) or mα6mß4* (∼ 2-4-fold)-nAChRs. EC50 values for nicotine acting at mα6mß4*-nAChR were unaffected by ß3 subunit residue substitutions in loop C or E. Thus, amino acid residues located in primary (loop C) or complementary (loops ß2-ß3 and E) interfaces of ß3 subunits are some of the molecular impediments for functional expression of mα6mß2ß3- or mα6mß4ß3-nAChRs.


Subject(s)
Amino Acids/chemistry , Protein Subunits/chemistry , Receptors, Nicotinic/chemistry , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Amino Acids/metabolism , Animals , Atropine/metabolism , Atropine/pharmacology , Female , Gene Expression , Humans , Membrane Potentials/drug effects , Mice , Molecular Sequence Data , Nicotine/metabolism , Nicotine/pharmacology , Oocysts/cytology , Oocysts/drug effects , Oocysts/metabolism , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Xenopus laevis
16.
Fish Shellfish Immunol ; 40(1): 288-95, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25038282

ABSTRACT

Ferritin, a major iron storage protein in most living organisms, plays a crucial role in iron metabolism. In this study, the ferritin subunit MnFer was identified in the oriental river prawn (Macrobrachium nipponense) and functionally characterized. The full-length cDNA of MnFer is 999 bp in size with a 122-bp 5'-untranslated region (UTR), a 364-bp 3'-UTR and a 513-bp open reading frame that encodes a protein possessing 171 amino acids and a deduced molecular weight of 19.40 kDa. Prawn ferritin transcripts are expressed in muscle, heart, hepatopancreas, gill, hemocytes, ovary and testis. Quantitative real-time PCR revealed that the abundance of ferritin transcript was highest in the hepatopancreas followed by muscle. Ferritin transcript expression in muscle increased six-fold 3 h after the injection of iron. In the gill, a four-fold increase in ferritin transcript expression was detected 3 h post-injection; the expression remained elevated for 48 h. Heart ferritin mRNA expression increased up to seven-fold at 24 h post-injection. No significant difference was found in the hepatopancreas. The iron binding capacity of recombinant ferritin protein was also demonstrated in this study. In hemocyte experiments, the transcriptional expression of MnFer showed the strongest response to Aeromonas hydrophila. As a whole, our study suggested that the ferritin from M. nipponense may play critical roles in cellular and organismic iron homeostasis along with in innate immune defense.


Subject(s)
Aeromonas hydrophila/physiology , Ferritins/genetics , Iron/pharmacology , Palaemonidae/genetics , Palaemonidae/immunology , Amino Acid Sequence , Animals , Arthropod Proteins/chemistry , Arthropod Proteins/genetics , Arthropod Proteins/metabolism , Base Sequence , DNA, Complementary/genetics , DNA, Complementary/metabolism , Ferritins/chemistry , Ferritins/metabolism , Molecular Sequence Data , Palaemonidae/metabolism , Phylogeny , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Real-Time Polymerase Chain Reaction , Sequence Alignment , Tissue Distribution
17.
Mol Brain ; 7: 35, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24886653

ABSTRACT

BACKGROUND: Functional heterologous expression of naturally-expressed and apparently functional mammalian α6*-nicotinic acetylcholine receptors (nAChRs; where '*' indicates presence of additional subunits) has been difficult. Here we wanted to investigate the role of N-terminal domain (NTD) residues of human (h) nAChR α6 subunit in the functional expression of hα6*-nAChRs. To this end, instead of adopting random mutagenesis as a tool, we used 15 NTD rare variations (i.e., Ser43Pro, Asn46Lys, Asp57Asn, Arg87Cys, Asp92Glu, Arg96His, Glu101Lys, Ala112Val, Ser156Arg, Asn171Lys, Ala184Asp, Asp199Tyr, Asn203Thr, Ile226Thr and Ser233Cys) in nAChR hα6 subunit to probe for their effect on the functional expression of hα6*-nAChRs. RESULTS: N-terminal α-helix (Asp57); complementary face/inner ß-fold (Arg87 or Asp92) and principal face/outer ß-fold (Ser156 or Asn171) residues in the hα6 subunit are crucial for functional expression of the hα6*-nAChRs as variations in these residues reduce or abrogate the function of hα6hß2*-, hα6hß4- and hα6hß4hß3-nAChRs. While variations at residues Ser43 or Asn46 (both in N-terminal α-helix) in hα6 subunit reduce hα6hß2*-nAChRs function those at residues Arg96 (ß2-ß3 loop), Asp199 (loop F) or Ser233 (ß10-strand) increase hα6hß2*-nAChR function. Similarly substitution of NTD α-helix (Asn46), loop F (Asp199), loop A (Ala112), loop B (Ala184), or loop C (Ile226) residues in hα6 subunit increase the function of hα6hß4-nAChRs. All other variations in hα6 subunit do not affect the function of hα6hß2*- and hα6hß4*-nAChRs. Incorporation of nAChR hß3 subunits always increase the function of wild-type or variant hα6hß4-nAChRs except for those of hα6(D57N, S156R, R87C or N171K)hß4-nAChRs. It appears Asp57Lys, Ser156Arg or Asn171Lys variations in hα6 subunit drive the hα6hß4hß3-nAChRs into a nonfunctional state as at spontaneously open hα6(D57N, S156R or N171K)hß4hß3V9'S-nAChRs (V9'S; transmembrane II 9' valine-to-serine mutation) agonists act as antagonists. Agonist sensitivity of hα6hß4- and/or hα6hß4hß3-nAChRs is nominally increased due to Arg96His, Ala184Asp, Asp199Tyr or Ser233Cys variation in hα6 subunit. CONCLUSIONS: Hence investigating functional consequences of natural variations in nAChR hα6 subunit we have discovered additional bases for cell surface functional expression of various subtypes of hα6*-nAChRs. Variations (Asp57Asn, Arg87Cys, Asp92Glu, Ser156Arg or Asn171Lys) in hα6 subunit that compromise hα6*-nAChR function are expected to contribute to individual differences in responses to smoked nicotine.


Subject(s)
Amino Acids/metabolism , Genetic Variation , Protein Subunits/chemistry , Protein Subunits/genetics , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/genetics , Acetylcholine/pharmacology , Amino Acid Sequence , Animals , Codon/genetics , Computational Biology , Humans , Ion Channel Gating/drug effects , Molecular Sequence Data , Mutant Proteins/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/metabolism , Receptors, Nicotinic/metabolism , Structure-Activity Relationship , Xenopus
18.
Bioorg Med Chem ; 22(4): 1276-84, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24462176

ABSTRACT

A dichloromethane extract of stems and roots of Pholidota chinensis (Orchidaceae) enhanced GABA-induced chloride currents (I(GABA)) by 132.75 ± 36.69% when tested at 100 µg/mL in a two-microelectrode voltage clamp assay, on Xenopus laevis oocytes expressing recombinant α1ß2γ2S GABA(A) receptors. By means of an HPLC-based activity profiling approach, the three structurally related stilbenoids coelonin (1), batatasin III (2), and pholidotol D (3) were identified in the active fractions of the extract. Dihydrostilbene 2 enhanced I(GABA) by 1512.19 ± 176.47% at 300 µM, with an EC50 of 52.51 ± 16.96 µM, while compounds 1 and 3 showed much lower activity. The relevance of conformational flexibility for receptor modulation by stilbenoids was confirmed with a series of 13 commercially available stilbenes and their corresponding semisynthetic dihydro derivatives. Dihydrostilbenes showed higher activity in the oocyte assay than their corresponding stilbenes. The dihydro derivatives of tetramethoxy-piceatannol (12) and pterostilbene (20) were the most active among these derivatives, but they showed lower efficiencies than compound 2. Batatasin III (2) showed high efficiency but no significant subunit specificity when tested on the receptor subtypes α1ß2γ2s, α2ß2γ2s, α3ß2γ2s, α4ß2γ2s, α5ß2γ2s, α1ß1γ2s, and α1ß3γ2s. Dihydrostilbenes represent a new scaffold for GABA(A) receptor modulators.


Subject(s)
Orchidaceae/chemistry , Receptors, GABA-A/chemistry , Stilbenes/chemistry , Animals , Oocytes/drug effects , Oocytes/metabolism , Orchidaceae/metabolism , Patch-Clamp Techniques , Plant Extracts/chemistry , Plant Roots/chemistry , Plant Roots/metabolism , Plant Stems/chemistry , Plant Stems/metabolism , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Stilbenes/isolation & purification , Stilbenes/pharmacology , Xenopus laevis/growth & development
19.
Plant Cell Environ ; 37(5): 1144-58, 2014 May.
Article in English | MEDLINE | ID: mdl-24237159

ABSTRACT

Polygalacturonase (PG), one of the hydrolases responsible for cell wall pectin degradation, is involved in organ consenescence and biotic stress in plants. PG1 is composed of a catalytic subunit, PG2, and a non-catalytic PG1ß subunit. OsBURP16 belongs to the PG1ß-like subfamily of BURP-family genes and encodes one putative PG1ß subunit precursor in rice (Oryza sativa L.). Transcription of OsBURP16 is induced by cold, salinity and drought stresses, as well as by abscisic acid (ABA) treatment. Analysis of plant survival rates, relative ion leakage rates, accumulation levels of H2 O2 and water loss rates of leaves showed that overexpression of OsBURP16 enhanced sensitivity to cold, salinity and drought stresses compared with controls. Young leaves of Ubi::OsBURP16 transgenic plants showed reduced cell adhesion and increased cuticular transpiration rate. Mechanical strength measurement of Ubi::OsBURP16 plants showed that reduced force was required to break leaves as compared with wild type. Transgenic rice showed enhanced PG activity and reduced pectin content. All these results suggested that overexpression of OsBURP16 caused pectin degradation and affected cell wall integrity as well as transpiration rate, which decreased tolerance to abiotic stresses.


Subject(s)
Oryza/cytology , Oryza/enzymology , Pectins/metabolism , Plant Proteins/metabolism , Polygalacturonase/metabolism , Stress, Physiological , Adaptation, Physiological/drug effects , Adaptation, Physiological/genetics , Amino Acid Sequence , Cell Adhesion/drug effects , Cell Wall/drug effects , Cell Wall/metabolism , Cold Temperature , Droughts , Enzyme Induction/drug effects , Gene Expression Regulation, Plant/drug effects , Molecular Sequence Data , Oryza/genetics , Oryza/physiology , Phenotype , Plant Leaves/drug effects , Plant Leaves/genetics , Plant Leaves/physiology , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Transpiration/drug effects , Plants, Genetically Modified , Polygalacturonase/chemistry , Polygalacturonase/genetics , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Sodium Chloride/pharmacology , Stress, Physiological/drug effects , Stress, Physiological/genetics , Uronic Acids/metabolism
20.
Structure ; 22(1): 149-55, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24268646

ABSTRACT

Structural studies of G protein-coupled receptors (GPCRs) extensively use the insertion of globular soluble protein domains to facilitate their crystallization. However, when inserted in the third intracellular loop (i3 loop), the soluble protein domain disrupts their coupling to G proteins and impedes the GPCRs functional characterization by standard G protein-based assays. Therefore, activity tests of crystallization-optimized GPCRs are essentially limited to their ligand binding properties using radioligand binding assays. Functional characterization of additional thermostabilizing mutations requires the insertion of similar mutations in the wild-type receptor to allow G protein-activation tests. We demonstrate that ion channel-coupled receptor technology is a complementary approach for a comprehensive functional characterization of crystallization-optimized GPCRs and potentially of any engineered GPCR. Ligand-induced conformational changes of the GPCRs are translated into electrical signal and detected by simple current recordings, even though binding of G proteins is sterically blocked by the added soluble protein domain.


Subject(s)
Biological Assay , Oocytes/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Protein Subunits/chemistry , Receptor, Muscarinic M2/chemistry , Recombinant Fusion Proteins/chemistry , Animals , Bacteriophage T4/chemistry , Bacteriophage T4/enzymology , Gene Expression , Genes, Reporter , Humans , Membrane Potentials/physiology , Mice , Muramidase/genetics , Muramidase/metabolism , Oocytes/cytology , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Protein Engineering , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism , Receptor, Muscarinic M2/genetics , Receptor, Muscarinic M2/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL