Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Pharmacol ; 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39044481

ABSTRACT

BACKGROUND AND PURPOSE: Allosterism is a regulatory mechanism for GPCRs that can be attained by ligand-binding or protein-protein interactions with another GPCR. We have studied the influence of the dimer interface on the allosteric properties of the A2A receptor and CB2 receptor heteromer. EXPERIMENTAL APPROACH: We have evaluated cAMP production, phosphorylation of signal-regulated kinases (pERK1/2), label-free dynamic mass redistribution, ß-arrestin 2 recruitment and bimolecular fluorescence complementation assays in the absence and presence of synthetic peptides that disrupt the formation of the heteromer. Molecular dynamic simulations provided converging evidence that the heteromeric interface influences the allosteric properties of the A2AR-CB2R heteromer. KEY RESULTS: Apo A2AR blocks agonist-induced signalling of CB2R. The disruptive peptides, with the amino acid sequence of transmembrane (TM) 6 of A2AR or CB2R, facilitate CB2R activation, suggesting that A2AR allosterically prevents the outward movement of TM 6 of CB2R for G protein binding. Significantly, binding of the selective antagonist SCH 58261 to A2AR also facilitated agonist-induced activation of CB2R. CONCLUSIONS AND IMPLICATIONS: It is proposed that the A2AR-CB2R heteromer contains distinct dimerization interfaces that govern its functional properties. The molecular interface between protomers of the A2AR-CB2R heteromer interconverted from TM 6 for apo or agonist-bound A2AR, blocking CB2R activation, to mainly the TM 1/7 interface for antagonist-bound A2AR, facilitating the independent opening of intracellular cavities for G protein binding. These novel results shed light on a different type of allosteric mechanism and extend the repertoire of GPCR heteromer signalling.

2.
Mol Microbiol ; 121(5): 850-864, 2024 05.
Article in English | MEDLINE | ID: mdl-38323722

ABSTRACT

The diarrheal disease cholera is caused by the versatile and responsive bacterium Vibrio cholerae, which is capable of adapting to environmental changes. Among others, the alternative sigma factor RpoS activates response pathways, including regulation of motility- and chemotaxis-related genes under nutrient-poor conditions in V. cholerae. Although RpoS has been well characterised, links between RpoS and other regulatory networks remain unclear. In this study, we identified the ArcAB two-component system to control rpoS transcription and RpoS protein stability in V. cholerae. In a manner similar to that seen in Escherichia coli, the ArcB kinase not only activates the response regulator ArcA but also RssB, the anti-sigma factor of RpoS. Our results demonstrated that, in V. cholerae, RssB is phosphorylated by ArcB, which subsequently activates RpoS proteolysis. Furthermore, ArcA acts as a repressor of rpoS transcription. Additionally, we determined that the cysteine residue at position 180 of ArcB is crucial for signal recognition and activity. Thus, our findings provide evidence linking RpoS response to the anoxic redox control system ArcAB in V. cholerae.


Subject(s)
Bacterial Proteins , Gene Expression Regulation, Bacterial , Sigma Factor , Vibrio cholerae , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Chemotaxis/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Gene Regulatory Networks , Phosphorylation , Repressor Proteins/metabolism , Repressor Proteins/genetics , Sigma Factor/metabolism , Sigma Factor/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Vibrio cholerae/genetics , Vibrio cholerae/metabolism
3.
Gut Microbes ; 15(2): 2281016, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37982663

ABSTRACT

In many Vibrio species, virulence is regulated by quorum sensing, which is regulated by a complex, multichannel, two-component phosphorelay circuit. Through this circuit, sensor kinases transmit sensory information to the phosphotransferase LuxU via a phosphotransfer mechanism, which in turn transmits the signal to the response regulator LuxO. For Vibrio parahaemolyticus, type III secretion system 1 (T3SS1) is required for cytotoxicity, but it is unclear how quorum sensing regulates T3SS1 expression. Herein, we report that a hybrid histidine kinase, ArcB, instead of LuxU, and sensor kinase LuxQ and response regulator LuxO, collectively orchestrate T3SS1 expression in V. parahaemolyticus. Under high oxygen conditions, LuxQ can interact with ArcB directly and phosphorylates the Hpt domain of ArcB. The Hpt domain of ArcB phosphorylates the downstream response regulator LuxO instead of ArcA. LuxO then activates transcription of the T3SS1 gene cluster. Under hypoxic conditions, ArcB autophosphorylates and phosphorylates ArcA, whereas ArcA does not participate in regulating the expression of T3SS1. Our data provides evidence of an alternative regulatory path involving the quorum sensing phosphorelay and adds another layer of understanding about the environmental regulation of gene expression in V. parahaemolyticus.


Subject(s)
Gastrointestinal Microbiome , Vibrio parahaemolyticus , Vibrio parahaemolyticus/genetics , Vibrio parahaemolyticus/metabolism , Quorum Sensing/genetics , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism , Bacterial Proteins/metabolism , Phosphotransferases/genetics , Gene Expression Regulation, Bacterial
4.
PeerJ ; 11: e16309, 2023.
Article in English | MEDLINE | ID: mdl-37849831

ABSTRACT

The complex metabolism of Escherichia coli has been extensively studied, including its response to oxygen availability. The ArcA/B two-component system (TCS) is the key regulator for the transition between these two environmental conditions and has been thoroughly characterized using genetic and biochemical approaches. Still, to date, limited structural data is available. The breakthrough provided by AlphaFold2 in 2021 has brought a reliable tool to the scientific community for assessing the structural features of complex proteins. In this report, we analyzed the structural aspects of the ArcA/B TCS using AlphaFold2 models. The models are consistent with the experimentally determined structures of ArcB kinase. The predicted structure of the dimeric form of ArcB is consistent with the extensive genetic and biochemical data available regarding mechanistic signal perception and regulation. The predicted interaction of the dimeric form of ArcB with its cognate response regulator (ArcA) is also consistent with both the forward and reverse phosphotransfer mechanisms. The ArcB model was used to detect putative binding cavities to anaerobic metabolites, encouraging testing of these predictions experimentally. Finally, the highly accurate models of other ArcB homologs suggest that different experimental approaches are needed to determine signal perception in kinases lacking the PAS domain. Overall, ArcB is a kinase with features that need further testing, especially in determining its crystal structure under different conditions.


Subject(s)
Escherichia coli Proteins , Anaerobiosis , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Dimerization , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Models, Theoretical , Phosphorylation , Protein Kinases/genetics , Repressor Proteins/genetics
5.
Res Microbiol ; 171(8): 301-310, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32721518

ABSTRACT

Aerobically growing Escherichia coli generates superoxide flux into the periplasm via the oxidation of dihydromenaquinone and simultaneously carries out continuous transmembrane cycling of glutathione (GSH). Here we have shown that, under the conditions of a gradual decrease in dissolved oxygen (dO2), characteristic of batch culture, the global regulatory system ArcB/ArcA can play an important role in the coordinated control of extracellular superoxide and GSH fluxes and their interaction with intracellular antioxidant systems. The lowest superoxide production was observed in the menA and arcB mutants, while the atpA, atpC and atpE mutants generated superoxide 1.3-1.5 times faster than the parent. The share of exported glutathione in the ubiC, atpA, atpC, and atpE mutants was 2-3 times higher compared to the parent. A high direct correlation (r = 0.87, p = 0.01) between extracellular superoxide and GSH was revealed. The menA and arcB mutants, as well as the cydD mutant lacking the GSH export system CydDC, were not capable of GSH excretion with a decrease in dO2, which indicates a positive control of GSH export by ArcB. In contrast, ArcB downregulates sodA, therefore, an inverse correlation (r = -0.86, p = 0.013) between superoxide production and sodA expression was observed.


Subject(s)
Escherichia coli/genetics , Escherichia coli/metabolism , Glutathione/metabolism , Superoxides/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Batch Cell Culture Techniques , Escherichia coli/growth & development , Gene Expression Regulation, Bacterial , Mutation , Oxidation-Reduction , Oxygen/metabolism , Signal Transduction
6.
Mol Cell ; 74(3): 481-493.e6, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30904393

ABSTRACT

The use of alternative translation initiation sites enables production of more than one protein from a single gene, thereby expanding the cellular proteome. Although several such examples have been serendipitously found in bacteria, genome-wide mapping of alternative translation start sites has been unattainable. We found that the antibiotic retapamulin specifically arrests initiating ribosomes at start codons of the genes. Retapamulin-enhanced Ribo-seq analysis (Ribo-RET) not only allowed mapping of conventional initiation sites at the beginning of the genes, but strikingly, it also revealed putative internal start sites in a number of Escherichia coli genes. Experiments demonstrated that the internal start codons can be recognized by the ribosomes and direct translation initiation in vitro and in vivo. Proteins, whose synthesis is initiated at internal in-frame and out-of-frame start sites, can be functionally important and contribute to the "alternative" bacterial proteome. The internal start sites may also play regulatory roles in gene expression.


Subject(s)
Genome, Bacterial/genetics , Peptide Chain Initiation, Translational , Proteome/genetics , Proteomics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Codon, Initiator/genetics , Diterpenes/pharmacology , Escherichia coli/drug effects , Escherichia coli/genetics , Gene Expression Regulation, Bacterial/drug effects , Genome, Bacterial/drug effects , RNA, Messenger/genetics , Ribosomes/drug effects , Ribosomes/genetics
7.
J Biol Chem ; 293(34): 13214-13223, 2018 08 24.
Article in English | MEDLINE | ID: mdl-29945971

ABSTRACT

The Arc (anoxic redox control) two-component system of Escherichia coli, comprising ArcA as the response regulator and ArcB as the sensor histidine kinase, modulates the expression of numerous genes in response to respiratory growth conditions. Under reducing growth conditions, ArcB autophosphorylates at the expense of ATP, and transphosphorylates ArcA via a His292 → Asp576 → His717 → Asp54 phosphorelay, whereas under oxidizing growth conditions, ArcB catalyzes the dephosphorylation of ArcA-P by a reverse Asp54 → His717 → Asp576 → Pi phosphorelay. However, the exact phosphoryl group transfer routes and the molecular mechanisms determining their directions are unclear. Here, we show that, during signal propagation, the His292 → Asp576 and Asp576 → His717 phosphoryl group transfers within ArcB dimers occur intra- and intermolecularly, respectively. Moreover, we report that, during signal decay, the phosphoryl group transfer from His717 to Asp576 takes place intramolecularly. In conclusion, we present a mechanism that dictates the direction of the phosphoryl group transfer within ArcB dimers and that enables the discrimination of the kinase and phosphatase activities of ArcB.


Subject(s)
Aspartic Acid/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Histidine/metabolism , Membrane Proteins/metabolism , Mutation , Protein Kinases/metabolism , Aspartic Acid/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Histidine/chemistry , Membrane Proteins/chemistry , Membrane Proteins/genetics , Phosphorylation , Protein Kinases/chemistry , Protein Kinases/genetics , Signal Transduction
8.
Antonie Van Leeuwenhoek ; 111(4): 609-617, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29139003

ABSTRACT

ArcAB is a two-component regulatory system that can help bacteria respond to and survive in a changing environment. To identify the function of ArcAB homologues in Serratia marcescens FS14, in-frame deletion mutants of the arcA, arcB and arcAB genes were constructed. Surprisingly, ArcB affects the motility of FS14, but ArcA does not. These results are the reverse of those found in Escherichia coli. Further studies demonstrated that ArcB could promote bacterial motility by activating the synthesis of flagella and particularly by activating the expression of the biosurfactant serrawettin W1. Our results suggest that ArcB may regulate FS14 motility by interacting with an unidentified response regulator other than ArcA. The regulation of ArcAB may be bacterial strain-specific, and the same regulatory system may participate in different mechanisms to adapt to different environments.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Serratia marcescens/genetics , Serratia marcescens/metabolism , Adaptation, Physiological/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Depsipeptides/genetics , Flagella/genetics , Flagella/metabolism , Gene Deletion , Hydrogen Peroxide/metabolism , Movement , Oxidative Stress , Promoter Regions, Genetic/genetics , Succinate Dehydrogenase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL