Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2741: 255-272, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38217658

RESUMEN

Bacterial small RNAs (sRNAs) can be equipped at the 5' end with triphosphate (5'PPP) or monophosphate (5'P) groups, depending on whether they are primary transcripts, undergo dephosphorylation or originate via processing. Often, 5' groups hallmark RNAs for rapid decay, but whether this also applies to sRNAs is little explored. Moreover, the sRNA 5'P group could activate endoribonuclease RNase E to cleave the base-paired target RNA, but a tool for investigation in vivo was lacking. Here, we describe a two-plasmid system suitable for the generation of 5' monophosphorylated RNAs on demand inside the cell. The sRNA gene of interest is fused to the 3' end of a fragment of sRNA GlmZ and transcribed from a plasmid in an IPTG-inducible manner. The fusion RNA gets cleaved upon arabinose-controlled expression of rapZ, provided on a compatible plasmid. Adaptor protein RapZ binds the GlmZ aptamer and directs RNase E to release the sRNA of choice with 5'P ends. An isogenic plasmid generating the same sRNA with a 5'PPP end allows for direct comparison. The fates of the sRNA variants and target RNA(s) are monitored by Northern blotting. This tool is applicable to E. coli and likely other enteric bacteria.


Asunto(s)
Proteínas de Escherichia coli , ARN Pequeño no Traducido , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Fosforilación , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , Estabilidad del ARN , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Regulación Bacteriana de la Expresión Génica
2.
RNA ; 26(9): 1198-1215, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32424019

RESUMEN

In Escherichia coli, endoribonuclease RNase E initiates degradation of many RNAs and represents a hub for post-transcriptional regulation. The tetrameric adaptor protein RapZ targets the small regulatory RNA GlmZ to degradation by RNase E. RapZ binds GlmZ through a domain located at the carboxyl terminus and interacts with RNase E, promoting GlmZ cleavage in the base-pairing region. When necessary, cleavage of GlmZ is counteracted by the homologous small RNA GlmY, which sequesters RapZ through molecular mimicry. In the current study, we addressed the molecular mechanism employed by RapZ. We show that RapZ mutants impaired in RNA-binding but proficient in binding RNase E are able to stimulate GlmZ cleavage in vivo and in vitro when provided at increased concentrations. In contrast, a truncated RapZ variant retaining RNA-binding activity but incapable of contacting RNase E lacks this activity. In agreement, we find that tetrameric RapZ binds the likewise tetrameric RNase E through direct interaction with its large globular domain within the catalytic amino terminus, independent of RNA. Although RapZ stimulates cleavage of at least one non-cognate RNA by RNase E in vitro, its activity is restricted to GlmZ in vivo as revealed by RNA sequencing, suggesting that certain features within the RNA substrate are also required for cleavage. In conclusion, RapZ boosts RNase E activity through interaction with its catalytic domain, which represents a novel mechanism of RNase E activation. In contrast, RNA-binding has a recruiting role, increasing the likelihood that productive RapZ/GlmZ/RNase E complexes form.


Asunto(s)
Endorribonucleasas/genética , Proteínas de Escherichia coli/genética , Mapas de Interacción de Proteínas/genética , ARN Pequeño no Traducido/genética , Proteínas de Unión al ARN/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Emparejamiento Base/genética , Catálisis , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica/genética , Imitación Molecular/genética , Estabilidad del ARN/genética
3.
EMBO J ; 39(6): e103848, 2020 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-32065419

RESUMEN

The RNA-binding protein RapZ cooperates with small RNAs (sRNAs) GlmY and GlmZ to regulate the glmS mRNA in Escherichia coli. Enzyme GlmS synthesizes glucosamine-6-phosphate (GlcN6P), initiating cell envelope biosynthesis. GlmZ activates glmS expression by base-pairing. When GlcN6P is ample, GlmZ is bound by RapZ and degraded through ribonuclease recruitment. Upon GlcN6P depletion, the decoy sRNA GlmY accumulates through a previously unknown mechanism and sequesters RapZ, suppressing GlmZ decay. This circuit ensures GlcN6P homeostasis and thereby envelope integrity. In this work, we identify RapZ as GlcN6P receptor. GlcN6P-free RapZ stimulates phosphorylation of the two-component system QseE/QseF by interaction, which in turn activates glmY expression. Elevated GlmY levels sequester RapZ into stable complexes, which prevents GlmZ decay, promoting glmS expression. Binding of GlmY also prevents RapZ from activating QseE/QseF, generating a negative feedback loop limiting the response. When GlcN6P is replenished, GlmY is released from RapZ and rapidly degraded. We reveal a multifunctional sRNA-binding protein that dynamically engages into higher-order complexes for metabolite signaling.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Glucosamina/análogos & derivados , Glucosa-6-Fosfato/análogos & derivados , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Escherichia coli/fisiología , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Glucosamina/metabolismo , Glucosa-6-Fosfato/metabolismo , ARN Bacteriano/genética , ARN Mensajero/genética , ARN Pequeño no Traducido/genética , Proteínas de Unión al ARN/genética , Receptores Adrenérgicos/genética , Receptores Adrenérgicos/metabolismo
4.
PLoS Genet ; 14(7): e1007547, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30040820

RESUMEN

Histidine kinase QseE and response regulator QseF compose a two-component system in Enterobacteriaceae. In Escherichia coli K-12 QseF activates transcription of glmY and of rpoE from Sigma 54-dependent promoters by binding to upstream activating sequences. Small RNA GlmY and RpoE (Sigma 24) are important regulators of cell envelope homeostasis. In pathogenic Enterobacteriaceae QseE/QseF are required for virulence. In enterohemorrhagic E. coli QseE was reported to sense the host hormone epinephrine and to regulate virulence genes post-transcriptionally through employment of GlmY. The qseEGF operon contains a third gene, qseG, which encodes a lipoprotein attached to the inner leaflet of the outer membrane. Here, we show that QseG is essential and limiting for activity of QseE/QseF in E. coli K-12. Metabolic 32P-labelling followed by pull-down demonstrates that phosphorylation of the receiver domain of QseF in vivo requires QseE as well as QseG. Accordingly, QseG acts upstream and through QseE/QseF by stimulating activity of kinase QseE. 32P-labelling also reveals an additional phosphorylation in the QseF C-terminus of unknown origin, presumably at threonine/serine residue(s). Pulldown and two-hybrid assays demonstrate interaction of QseG with the periplasmic loop of QseE. A mutational screen identifies the Ser58Asn exchange in the periplasmic loop of QseE, which decreases interaction with QseG and concomitantly lowers QseE/QseF activity, indicating that QseG activates QseE by interaction. Finally, epinephrine is shown to have a moderate impact on QseE activity in E. coli K-12. Epinephrine slightly stimulates QseF phosphorylation and thereby glmY transcription, but exclusively during stationary growth and this requires both, QseE and QseG. Our data reveal a three-component signaling system, in which the phosphorylation state of QseE/QseF is governed by interaction with lipoprotein QseG in response to a signal likely derived from the cell envelope.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de Unión al ADN/metabolismo , Escherichia coli K12/fisiología , Proteínas de Escherichia coli/metabolismo , Periplasma/metabolismo , Receptores Adrenérgicos/metabolismo , Epinefrina/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Operón/genética , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Regiones Promotoras Genéticas/genética , Unión Proteica/fisiología , Transcripción Genética/efectos de los fármacos
5.
Microbiol Spectr ; 6(2)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29573258

RESUMEN

Survival of bacteria in ever-changing habitats with fluctuating nutrient supplies requires rapid adaptation of their metabolic capabilities. To this end, carbohydrate metabolism is governed by complex regulatory networks including posttranscriptional mechanisms that involve small regulatory RNAs (sRNAs) and RNA-binding proteins. sRNAs limit the response to substrate availability and set the threshold or time required for induction and repression of carbohydrate utilization systems. Carbon catabolite repression (CCR) also involves sRNAs. In Enterobacteriaceae, sRNA Spot 42 cooperates with the transcriptional regulator cyclic AMP (cAMP)-receptor protein (CRP) to repress secondary carbohydrate utilization genes when a preferred sugar is consumed. In pseudomonads, CCR operates entirely at the posttranscriptional level, involving RNA-binding protein Hfq and decoy sRNA CrcZ. Moreover, sRNAs coordinate fluxes through central carbohydrate metabolic pathways with carbohydrate availability. In Gram-negative bacteria, the interplay between RNA-binding protein CsrA and its cognate sRNAs regulates glycolysis and gluconeogenesis in response to signals derived from metabolism. Spot 42 and cAMP-CRP jointly downregulate tricarboxylic acid cycle activity when glycolytic carbon sources are ample. In addition, bacteria use sRNAs to reprogram carbohydrate metabolism in response to anaerobiosis and iron limitation. Finally, sRNAs also provide homeostasis of essential anabolic pathways, as exemplified by the hexosamine pathway providing cell envelope precursors. In this review, we discuss the manifold roles of bacterial sRNAs in regulation of carbon source uptake and utilization, substrate prioritization, and metabolism.


Asunto(s)
Bacterias/metabolismo , Metabolismo de los Hidratos de Carbono/fisiología , ARN Pequeño no Traducido/metabolismo , Azúcares/metabolismo , Bacterias/genética , Represión Catabólica , Ciclo del Ácido Cítrico/fisiología , Proteína Receptora de AMP Cíclico/metabolismo , Regulación hacia Abajo , Enterobacteriaceae/metabolismo , Regulación Bacteriana de la Expresión Génica , Gluconeogénesis/fisiología , Glucólisis/fisiología , Hexosaminas/metabolismo , Homeostasis , Proteína de Factor 1 del Huésped/metabolismo , Redes y Vías Metabólicas , ARN Bacteriano/metabolismo , ARN Pequeño no Traducido/genética , Proteínas de Unión al ARN , Proteínas Represoras/metabolismo , Transducción de Señal , Transactivadores
6.
Mol Microbiol ; 106(1): 54-73, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28714556

RESUMEN

The two-component system KdpD/KdpE governs K+ homeostasis by controlling synthesis of the high affinity K+ transporter KdpFABC. When sensing low environmental K+ concentrations, the dimeric kinase KdpD autophosphorylates in trans and transfers the phosphoryl-group to the response regulator KdpE, which subsequently activates kdpFABC transcription. In Escherichia coli, KdpD can also be activated by interaction with the non-phosphorylated form of the accessory protein PtsN. PtsN stimulates KdpD kinase activity thereby increasing phospho-KdpE levels. Here, we analyzed the interplay between KdpD/KdpE and PtsN. PtsN binds specifically to the catalytic DHp domain of KdpD, which is also contacted by KdpE. Accordingly, PtsN and KdpE compete for binding, providing a paradox. Low levels of non-phosphorylated PtsN stimulate, whereas high amounts reduce kdpFABC expression by blocking access of KdpE to KdpD. Ligand fishing experiments provided insight as they revealed ternary complex formation of PtsN/KdpD2 /KdpE in vivo demonstrating that PtsN and KdpE bind different protomers in the KdpD dimer. PtsN may bind one protomer to stimulate phosphorylation of the second KdpD protomer, which then phosphorylates bound KdpE. Phosphorylation of PtsN prevents its incorporation in ternary complexes. Interaction with the conserved DHp domain enables PtsN to regulate additional kinases such as PhoR.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Proteínas Quinasas/metabolismo , Sitios de Unión , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica/genética , Histidina Quinasa , Proteínas de Transporte de Membrana/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Fosforilación , Potasio/metabolismo , Dominios Proteicos , Subunidades de Proteína/metabolismo , Transducción de Señal , Transactivadores/metabolismo
7.
Front Microbiol ; 7: 908, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27379045

RESUMEN

Formation of glucosamine-6-phosphate (GlcN6P) by enzyme GlcN6P synthase (GlmS) represents the first step in bacterial cell envelope synthesis. In Escherichia coli, expression of glmS is controlled by small RNAs (sRNAs) GlmY and GlmZ. GlmZ activates the glmS mRNA by base-pairing. When not required, GlmZ is bound by adapter protein RapZ and recruited to cleavage by RNase E inactivating the sRNA. The homologous sRNA GlmY activates glmS indirectly. When present at high levels, GlmY sequesters RapZ by an RNA mimicry mechanism suppressing cleavage of GlmZ. The interplay of both sRNAs is believed to adjust GlmS synthesis to the needs of the cell, i.e., to achieve GlcN6P homeostasis. Bacilysin (tetaine) and Nva-FMDP are dipeptide antibiotics that impair cell envelope synthesis by inhibition of enzyme GlmS through covalent modification. However, although taken up efficiently, these antibiotics are less active against E. coli for reasons unknown so far. Here we show that the GlmY/GlmZ circuit provides resistance. Inhibition of GlmS causes GlcN6P deprivation leading to activation of GlmY and GlmZ, which in turn trigger glmS overexpression in a dosage-dependent manner. Mutation of glmY or glmZ disables this response and renders the bacteria highly susceptible to GlmS inhibitors. Thus, E. coli compensates inhibition of GlmS by increasing its synthesis through the GlmY/GlmZ pathway. This mechanism is also operative in Salmonella indicating that it is conserved in Enterobacteriaceae possessing these sRNAs. As GlmY apparently responds to GlcN6P, co-application of a non-metabolizable GlcN6P analog may prevent activation of the sRNAs and thereby increase the bactericidal activity of GlmS inhibitors against wild-type bacteria. Initial experiments using glucosamine-6-sulfate support this possibility. Thus, GlcN6P analogs might be considered for co-application with GlmS inhibitors in combined therapy to treat infections caused by pathogenic Enterobacteriaceae.

8.
Nucleic Acids Res ; 44(2): 824-37, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26531825

RESUMEN

In E. coli, small RNA GlmZ activates the glmS mRNA by base-pairing in an Hfq dependent manner. When not required, GlmZ is bound by adaptor protein RapZ and recruited to RNase E, which cleaves GlmZ in its base-pairing sequence. Small RNA GlmY counteracts cleavage of GlmZ by sequestration of RapZ. Although both sRNAs are highly homologous, only GlmZ specifically binds Hfq and undergoes cleavage by RNase E. We used domain swapping to identify the responsible modules. Two elements, the 3' terminal oligo(U) stretch and the base-pairing region enable GlmZ to interact with Hfq. Accordingly, Hfq inhibits cleavage of GlmZ, directing it to base-pairing. Intriguingly, the central stem loop of GlmZ is decisive for cleavage, whereas the sequence comprising the actual cleavage site is dispensable. Assisted by RapZ, RNase E cleaves any RNA fused to the 3' end of this module. These results suggest a novel mode for RNase E recognition, in which one of the required handholds in the substrate is replaced by an RNA binding protein. This device can generate RNAs of interest in their 5' monophosphorylated form on demand. As these species are rapidly degraded, this tool allows to regulate gene expression post-transcriptionally by modulation of RapZ levels.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Endorribonucleasas/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteína de Factor 1 del Huésped/metabolismo , ARN Bacteriano/metabolismo , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/genética , Emparejamiento Base , Sitios de Unión , Endorribonucleasas/genética , Proteínas de Escherichia coli/genética , Proteína de Factor 1 del Huésped/genética , ARN Bacteriano/química , ARN Bacteriano/genética , ARN Mensajero/metabolismo , ARN Pequeño no Traducido/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
9.
J Mol Microbiol Biotechnol ; 25(2-3): 168-77, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26159077

RESUMEN

Many Proteobacteria possess the regulatory nitrogen-related phosphotransferase system (PTS(Ntr)), which operates in parallel to the transport PTS. PTS(Ntr) is composed of the proteins EI(Ntr) and NPr and the final phosphate acceptor EIIA(Ntr). Both PTSs can exchange phosphoryl groups among each other. Proteins governing K(+) uptake represent a major target of PTS(Ntr) in Escherichia coli. Nonphosphorylated EIIA(Ntr) binds and stimulates the K(+) sensor KdpD, which activates expression of the kdpFABC operon encoding a K(+) transporter. Here we show that this regulation also operates in an ilvG(+) strain ruling out previous concern about interference with a nonfunctional ilvG allele present in many strains. Furthermore, we analyzed phosphorylation of EIIA(Ntr). In wild-type cells EIIA(Ntr) is predominantly phosphorylated, regardless of the growth stage and the utilized carbon source. However, cross-phosphorylation of EIIA(Ntr) by the transport PTS becomes apparent in the absence of EI(Ntr): EIIA(Ntr) is predominantly nonphosphorylated when cells grow on a PTS sugar and phosphorylated when a non-PTS carbohydrate is utilized. These differences in phosphorylation are transduced into corresponding kdpFABC transcription levels. Thus, the transport PTS may affect phosphorylation of EIIA(Ntr) and accordingly modulate processes controlled by EIIA(Ntr). Our data suggest that this cross-talk becomes most relevant under conditions that would inhibit activity of EI(Ntr).


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Nitrógeno/metabolismo , Operón , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Fosfotransferasas/metabolismo , Potasio/metabolismo , Adenosina Trifosfatasas/genética , Proteínas de Transporte de Catión/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Fosforilación , Fosfotransferasas/genética
10.
Mol Microbiol ; 92(4): 641-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24707963

RESUMEN

Bacteria use intricately interconnected mechanisms acting at the transcriptional and post-transcriptional level to adjust gene expression to their needs. An intriguing example found in the chitosugar utilization systems of Escherichia coli and Salmonella is uncovered in a study by Plumbridge and colleagues. Three transcription factors (TFs), a small regulatory RNA (sRNA) and a sRNA trap cooperate to set thresholds and dynamics in regulation of chitosugar utilization. Specifically, under inducing conditions a decoy site on the polycistronic chitobiose (chbBCARFG) mRNA sequesters sRNA ChiX, which represses synthesis of the separately encoded chitoporin ChiP. Base-pairing of ChiX with its decoy has no role for the chb genes themselves when the mRNA is in excess. In the absence of substrate, however, this base-pairing tightly represses chbC encoding a subunit of the chitosugar transporter. Thus, one and the same sRNA/mRNA interaction serves different regulatory functions under different environmental conditions. The employment of RNA decoys to control the activities of post-transcriptional regulators themselves is an increasingly recognized mechanism in gene regulation. Another observation in the current study highlights the possibility that decoy sites might even exist on the DNA controlling the availability of TFs for their target promoters.


Asunto(s)
Quitina/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Salmonella/genética , Salmonella/metabolismo
11.
RNA Biol ; 11(5): 433-42, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24667238

RESUMEN

In Escherichia coli, small RNAs GlmY and GlmZ feedback control synthesis of glucosamine-6-phosphate (GlcN6P) synthase GlmS, a key enzyme required for synthesis of the cell envelope. Both small RNAs are highly similar, but only GlmZ is able to activate the glmS mRNA by base-pairing. Abundance of GlmZ is controlled at the level of decay by RNase adaptor protein RapZ. RapZ binds and targets GlmZ to degradation by RNase E via protein-protein interaction. GlmY activates glmS indirectly by protecting GlmZ from degradation. Upon GlcN6P depletion, GlmY accumulates and sequesters RapZ in an RNA mimicry mechanism, thus acting as an anti-adaptor. As a result, this regulatory circuit adjusts synthesis of GlmS to the level of its enzymatic product, thereby mediating GlcN6P homeostasis. The interplay of RNase adaptor proteins and anti-adaptors provides an elegant means how globally acting RNases can be re-programmed to cleave a specific transcript in response to a cognate stimulus.


Asunto(s)
Pared Celular/genética , Imitación Molecular , Procesamiento Postranscripcional del ARN , ARN Bacteriano/genética , ARN Pequeño no Traducido/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Emparejamiento Base , Pared Celular/metabolismo , Endorribonucleasas/metabolismo , Activación Enzimática , Regulación Bacteriana de la Expresión Génica , Glucosamina/análogos & derivados , Glucosamina/metabolismo , Glucosa-6-Fosfato/análogos & derivados , Glucosa-6-Fosfato/metabolismo , Bacterias Gramnegativas/genética , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/genética , Bacterias Grampositivas/metabolismo , Interacciones Huésped-Patógeno , Operón/genética , Poliadenilación , Regiones Promotoras Genéticas , Unión Proteica , Estabilidad del ARN , ARN Bacteriano/química , ARN Bacteriano/metabolismo , ARN Catalítico/genética , ARN Catalítico/metabolismo , ARN Pequeño no Traducido/química , ARN Pequeño no Traducido/metabolismo , Ribonucleasas/metabolismo
12.
Genes Dev ; 27(5): 552-64, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23475961

RESUMEN

Bacterial small RNAs (sRNAs) are well established to regulate diverse cellular processes, but how they themselves are regulated is less understood. Recently, we identified a regulatory circuit wherein the GlmY and GlmZ sRNAs of Escherichia coli act hierarchically to activate mRNA glmS, which encodes glucosamine-6-phosphate (GlcN6P) synthase. Although the two sRNAs are highly similar, only GlmZ is a direct activator that base-pairs with the glmS mRNA, aided by protein Hfq. GlmY, however, does not bind Hfq and activates glmS indirectly by protecting GlmZ from RNA cleavage. This complex regulation feedback controls the levels of GlmS protein in response to its product, GlcN6P, a key metabolite in cell wall biosynthesis. Here, we reveal the molecular basis for the regulated turnover of GlmZ, identifying RapZ (RNase adaptor protein for sRNA GlmZ; formerly YhbJ) as a novel type of RNA-binding protein that recruits the major endoribonuclease RNase E to GlmZ. This involves direct interaction of RapZ with the catalytic domain of RNase E. GlmY binds RapZ through a secondary structure shared by both sRNAs and therefore acts by molecular mimicry as a specific decoy for RapZ. Thus, in analogy to regulated proteolysis, RapZ is an adaptor, and GlmY is an anti-adaptor in regulated turnover of a regulatory small RNA.


Asunto(s)
Endorribonucleasas/metabolismo , Escherichia coli/enzimología , Estabilidad del ARN , ARN Bacteriano/metabolismo , Secuencia de Aminoácidos , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Imitación Molecular , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas de Unión al ARN , Alineación de Secuencia
13.
Artículo en Inglés | MEDLINE | ID: mdl-23385747

RESUMEN

The protein YhbJ from Escherichia coli was previously reported to be involved in the regulation of glucosamine-6-phosphate synthase (GlmS) synthesis. YhbJ controls a regulatory cascade composed of the two small RNAs GlmY and GlmZ, which in turn regulate GlmS synthesis. For structural characterization, YhbJ was cloned, expressed and purified to homogeneity by Strep-tag affinity chromatography and size-exclusion chromatography. Multi-angle laser light-scattering analysis revealed its homotrimeric state in solution. The protein crystallized in two distinct trigonal crystal forms, with unit-cell parameters a = b = 91.62, c = 352.82 Å for space group P321 and a = b = 92.72, c = 156.75 Å for one of the enantiomorphic space groups P3(1) or P3(2). Preliminary analysis of the diffraction data suggests the presence of approximately three to seven molecules per asymmetric unit. Owing to the lack of a suitable homologous model, structure determination by means of MIR and MAD methods is required.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , ARN Bacteriano/metabolismo , Cristalización , Cristalografía por Rayos X , Proteínas de Escherichia coli/química , Proteínas de Unión al ARN
14.
Mol Microbiol ; 86(1): 96-110, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22812494

RESUMEN

Many Proteobacteria possess the paralogous PTS(Ntr), in addition to the sugar transport phosphotransferase system (PTS). In the PTS(Ntr) phosphoryl-groups are transferred from phosphoenolpyruvate to protein EIIA(Ntr) via the phosphotransferases EI(Ntr) and NPr. The PTS(Ntr) has been implicated in regulation of diverse physiological processes. In Escherichia coli, the PTS(Ntr) plays a role in potassium homeostasis. In particular, EIIA(Ntr) binds to and stimulates activity of a two-component histidine kinase (KdpD) resulting in increased expression of the genes encoding the high-affinity K(+) transporter KdpFABC. Here, we show that the phosphate (pho) regulon is likewise modulated by PTS(Ntr). The pho regulon, which comprises more than 30 genes, is activated by the two-component system PhoR/PhoB under conditions of phosphate starvation. Mutants lacking EIIA(Ntr) are unable to fully activate the pho genes and exhibit a growth delay upon adaptation to phosphate limitation. In contrast, pho expression is increased above the wild-type level in mutants deficient for EIIA(Ntr) phosphorylation suggesting that non-phosphorylated EIIA(Ntr) modulates pho. Protein interaction analyses reveal binding of EIIA(Ntr) to histidine kinase PhoR. This interaction increases the amount of phosphorylated response regulator PhoB. Thus, EIIA(Ntr) is an accessory protein that modulates the activities of two distinct sensor kinases, KdpD and PhoR, in E. coli.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Mapeo de Interacción de Proteínas , Fusión Artificial Génica , Escherichia coli/crecimiento & desarrollo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Eliminación de Gen , Perfilación de la Expresión Génica , Genes Reporteros , Fosfatos/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , beta-Galactosidasa/análisis , beta-Galactosidasa/genética
15.
Curr Opin Microbiol ; 15(2): 132-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22197250

RESUMEN

Bacterial two-component systems (TCSs) and small regulatory RNAs (sRNAs) form densely interconnected networks that integrate and transduce information from the environment into fine-tuned changes of gene expression. Many TCSs control target genes indirectly through regulation of sRNAs, which in turn regulate gene expression by base-pairing with mRNAs or targeting a protein. Conversely, sRNAs may control TCS synthesis, thereby recruiting the TCS regulon to other regulatory networks. Several TCSs control expression of multiple homologous sRNAs providing the regulatory networks with further flexibility. These sRNAs act redundantly, additively or hierarchically on targets. The regulatory speed of sRNAs and their unique features in gene regulation make them ideal players extending the flexibility, dynamic range or timing of TCS signaling.


Asunto(s)
Bacterias/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , ARN Pequeño no Traducido/metabolismo , Transducción de Señal , Bacterias/metabolismo , Proteínas Bacterianas/genética , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Pequeño no Traducido/genética
16.
Nucleic Acids Res ; 39(4): 1294-309, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20965974

RESUMEN

Small RNAs GlmY and GlmZ compose a cascade that feedback-regulates synthesis of enzyme GlmS in Enterobacteriaceae. Here, we analyzed the transcriptional regulation of glmY/glmZ from Yersinia pseudotuberculosis, Salmonella typhimurium and Escherichia coli, as representatives for other enterobacterial species, which exhibit similar promoter architectures. The GlmY and GlmZ sRNAs of Y. pseudotuberculosis are transcribed from σ(54)-promoters that require activation by the response regulator GlrR through binding to three conserved sites located upstream of the promoters. This also applies to glmY/glmZ of S. typhimurium and glmY of E. coli, but as a difference additional σ(70)-promoters overlap the σ(54)-promoters and initiate transcription at the same site. In contrast, E. coli glmZ is transcribed from a single σ(70)-promoter. Thus, transcription of glmY and glmZ is controlled by σ(54) and the two-component system GlrR/GlrK (QseF/QseE) in Y. pseudotuberculosis and presumably in many other Enterobacteria. However, in a subset of species such as E. coli this relationship is partially lost in favor of σ(70)-dependent transcription. In addition, we show that activity of the σ(54)-promoter of E. coli glmY requires binding of the integration host factor to sites upstream of the promoter. Finally, evidence is provided that phosphorylation of GlrR increases its activity and thereby sRNA expression.


Asunto(s)
Enterobacteriaceae/genética , Regulación Bacteriana de la Expresión Génica , ARN Bacteriano/genética , ARN Pequeño no Traducido/genética , Transcripción Genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Proteínas de Unión al ADN/metabolismo , Escherichia coli/genética , Factores de Integración del Huésped/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa Sigma 54/metabolismo , ARN Bacteriano/metabolismo , ARN Pequeño no Traducido/metabolismo , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Sintenía , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/metabolismo
17.
Mol Microbiol ; 74(5): 1054-70, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19843219

RESUMEN

In Escherichia coli synthesis of glucosamine-6-phosphate synthase GlmS is feedback-controlled by a regulatory cascade composed of small RNAs GlmY and GlmZ. When GlcN6P becomes limiting, GlmY accumulates and inhibits processing of GlmZ. Full-length GlmZ base-pairs with the glmS transcript and activates synthesis of GlmS, which re-synthesizes GlcN6P. Here we show that glmY expression is controlled by two overlapping promoters with the same transcription start site. A sigma(70)-dependent promoter contributes to glmY transcription during exponential growth. Alternatively, glmY can be transcribed from a sigma(54)-dependent promoter, which requires the YfhK/YfhA two-component system for activity. YfhK is a sensor kinase and YfhA is a response regulator that contains a sigma(54) interaction domain. YfhA binds to a DNA region located more than 100 bp upstream of glmY. Three copies of the conserved sequence TGTCN(10)GACA contribute to binding, and the two sites next to glmY are essential for activation of the sigma(54)-dependent promoter by YfhA. YfhK and YfhA upregulate GlmY when cells enter the stationary growth phase, whereas regulation by glucosamine-6-phosphate occurs post GlmY transcription. Target genes regulated by YfhK and YfhA were unknown so far. We propose to rename these proteins to GlrK and GlrR, for glmY regulating kinase and response regulator respectively.


Asunto(s)
Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Regiones Promotoras Genéticas , ARN Bacteriano/genética , ARN no Traducido/genética , Transcripción Genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Genes Bacterianos , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/genética , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/metabolismo , Polinucleotido Adenililtransferasa/genética , Polinucleotido Adenililtransferasa/metabolismo , Proteínas Quinasas/metabolismo , ARN Polimerasa Sigma 54/metabolismo , ARN Bacteriano/metabolismo , ARN no Traducido/metabolismo , Factor sigma/metabolismo , Sitio de Iniciación de la Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA