Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sandrine Poncet is active.

Publication


Featured researches published by Sandrine Poncet.


The EMBO Journal | 2003

Transmembrane modulator-dependent bacterial tyrosine kinase activates UDP-glucose dehydrogenases

Ivan Mijakovic; Sandrine Poncet; Grégory Boël; Alain Mazé; Sylvie Gillet; Emmanuel Jamet; Paulette Decottignies; Christophe Grangeasse; Patricia Doublet; Pierre Le Maréchal; Josef Deutscher

Protein‐tyrosine kinases regulating bacterial exopolysaccharide synthesis autophosphorylate on tyrosines located in a conserved C‐terminal region. So far no other substrates have been identified for these kinases. Here we demonstrate that Bacillus subtilis YwqD not only autophosphorylates at Tyr‐228, but that it also phosphorylates the two UDP‐glucose dehydrogenases (UDP‐glucose DHs) YwqF and TuaD at a tyrosine residue. However, phosphorylation of YwqF and TuaD occurs only in the presence of the transmembrane protein YwqC. The presumed intracellular C‐terminal part of YwqC (last 50 amino acids) seems to interact with the tyrosine‐kinase and to allow YwqD‐catalysed phosphorylation of the two UDP‐glucose DHs, which are key enzymes for the synthesis of acidic polysaccharides. However, only when phosphorylated by YwqD do the two enzymes exhibit detectable UDP‐glucose DH activity. Dephosphorylation of P‐Tyr‐YwqF and P‐Tyr‐TuaD by the P‐Tyr‐protein phosphatase YwqE switched off their UDP‐glucose DH activity. YwqE, which is encoded by the fourth gene of the B.subtilis ywqCDEF operon, also dephosphorylates P‐Tyr‐YwqD.


Journal of Bacteriology | 2010

Complete Genome Sequence of the Probiotic Lactobacillus casei Strain BL23

Alain Mazé; Grégory Boël; Manuel Zúñiga; Alexa Bourand; Valentin Loux; María J. Yebra; Vicente Monedero; Karine Correia; Noémie Jacques; Sophie Beaufils; Sandrine Poncet; Philippe Joyet; Eliane Milohanic; Serge Casaregola; Yanick Auffray; Gaspar Pérez-Martínez; Jean-François Gibrat; Monique Zagorec; Christof Francke; Axel Hartke; Josef Deutscher

The entire genome of Lactobacillus casei BL23, a strain with probiotic properties, has been sequenced. The genomes of BL23 and the industrially used probiotic strain Shirota YIT 9029 (Yakult) seem to be very similar.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Pyrophosphate-producing protein dephosphorylation by HPr kinase/phosphorylase: A relic of early life?

Ivan Mijakovic; Sandrine Poncet; Anne Galinier; Vicente Monedero; Sonia Fieulaine; Joël Janin; Sylvie Nessler; José Antonio Marquez; Klaus Scheffzek; Sonja Hasenbein; Wolfgang Hengstenberg; Josef Deutscher

In most Gram-positive bacteria, serine-46-phosphorylated HPr (P-Ser-HPr) controls the expression of numerous catabolic genes (≈10% of their genome) by acting as catabolite corepressor. HPr kinase/phosphorylase (HprK/P), the bifunctional sensor enzyme for catabolite repression, phosphorylates HPr, a phosphocarrier protein of the sugar-transporting phosphoenolpyruvate/glycose phosphotransferase system, in the presence of ATP and fructose-1,6-bisphosphate but dephosphorylates P-Ser-HPr when phosphate prevails over ATP and fructose-1,6-bisphosphate. We demonstrate here that P-Ser-HPr dephosphorylation leads to the formation of HPr and pyrophosphate. HprK/P, which binds phosphate at the same site as the β phosphate of ATP, probably uses the inorganic phosphate to carry out a nucleophilic attack on the phosphoryl bond in P-Ser-HPr. HprK/P is the first enzyme known to catalyze P-protein dephosphorylation via this phospho–phosphorolysis mechanism. This reaction is reversible, and at elevated pyrophosphate concentrations, HprK/P can use pyrophosphate to phosphorylate HPr. Growth of Bacillus subtilis on glucose increased intracellular pyrophosphate to concentrations (≈6 mM), which in in vitro tests allowed efficient pyrophosphate-dependent HPr phosphorylation. To effectively dephosphorylate P-Ser-HPr when glucose is exhausted, the pyrophosphate concentration in the cells is lowered to 1 mM. In B. subtilis, this might be achieved by YvoE. This protein exhibits pyrophosphatase activity, and its gene is organized in an operon with hprK.


The EMBO Journal | 2001

Mutations lowering the phosphatase activity of HPr kinase/phosphatase switch off carbon metabolism.

Vicente Monedero; Sandrine Poncet; Ivan Mijakovic; Sonia Fieulaine; Valérie Dossonnet; Isabelle Martin-Verstraete; Sylvie Nessler; Josef Deutscher

The oligomeric bifunctional HPr kinase/P‐Ser‐HPr phosphatase (HprK/P) regulates many metabolic functions in Gram‐positive bacteria by phosphorylating the phosphocarrier protein HPr at Ser46. We isolated Lactobacillus casei hprK alleles encoding mutant HprK/Ps exhibiting strongly reduced phosphatase, but almost normal kinase activity. Two mutations affected the Walker motif A of HprK/P and four a conserved C‐terminal region in contact with the ATP‐binding site of an adjacent subunit in the hexamer. Kinase and phosphatase activity appeared to be closely associated and linked to the Walker motif A, but dephosphorylation of seryl‐phosphorylated HPr (P‐Ser‐HPr) is not simply a reversal of the kinase reaction. When the hprKV267F allele was expressed in Bacillus subtilis, the strongly reduced phosphatase activity of the mutant enzyme led to increased amounts of P‐Ser‐HPr. The hprK V267F mutant was unable to grow on carbohydrates transported by the phosphoenolpyruvate:glycose phosphotransferase system (PTS) and on most non‐PTS carbohydrates. Disrupting ccpA relieved the growth defect only on non‐PTS sugars, whereas replacing Ser46 in HPr with alanine also restored growth on PTS substrates.


The EMBO Journal | 2001

X-ray structure of HPr kinase: a bacterial protein kinase with a P-loop nucleotide-binding domain.

Sonia Fieulaine; Solange Moréra; Sandrine Poncet; Vicente Monedero; Virginie Gueguen-Chaignon; Anne Galinier; Joël Janin; Josef Deutscher; Sylvie Nessler

HPr kinase/phosphatase (HprK/P) is a key regulatory enzyme controlling carbon metabolism in Gram‐ positive bacteria. It catalyses the ATP‐dependent phosphorylation of Ser46 in HPr, a protein of the phosphotransferase system, and also its dephosphorylation. HprK/P is unrelated to eukaryotic protein kinases, but contains the Walker motif A characteristic of nucleotide‐binding proteins. We report here the X‐ray structure of an active fragment of Lactobacillus casei HprK/P at 2.8 Å resolution, solved by the multiwavelength anomalous dispersion method on a seleniated protein (PDB code 1jb1). The protein is a hexamer, with each subunit containing an ATP‐binding domain similar to nucleoside/nucleotide kinases, and a putative HPr‐binding domain unrelated to the substrate‐binding domains of other kinases. The Walker motif A forms a typical P‐loop which binds inorganic phosphate in the crystal. We modelled ATP binding by comparison with adenylate kinase, and designed a tentative model of the complex with HPr based on a docking simulation. The results confirm that HprK/P represents a new family of protein kinases, first identified in bacteria, but which may also have members in eukaryotes.


Molecular Microbiology | 2002

Antitermination by GlpP, catabolite repression via CcpA and inducer exclusion triggered by P~GlpK dephosphorylation control Bacillus subtilis glpFK expression

Emmanuelle Darbon; Pascale Servant; Sandrine Poncet; Josef Deutscher

The Bacillus subtilis glpFK operon encoding the glycerol transport facilitator (GlpF) and glycerol kinase (GlpK) is induced by glycerol‐3‐P and repressed by rapidly metabolizable sugars. Carbon catabolite repression (CCR) of glpFK is partly mediated via a catabolite response element cre preceding glpFK. This operator site is recognized by the catabolite control protein A (CcpA) in complex with one of its co‐repressors, P‐Ser‐HPr or P‐Ser‐Crh. HPr is a component of the phosphoenolpyruvate:sugar phos‐photransferase system (PTS), and Crh is an HPr homologue. The hprK‐encoded HPr kinase phosphorylates HPr and Crh at Ser‐46. But in neither ccpA nor hprK mutants was expression of a glpF′–lacZ fusion relieved from CCR, as a second, CcpA‐independent CCR mechanism implying the terminator tglpFK, whose formation is prevented by the glycerol‐3‐P‐activated antiterminator GlpP, is operative. Deletion of tglpFK led to elevated expression of the glpF′–lacZ fusion and to partial relief from CCR. CCR completely disappeared in ΔtglpFK mutants carrying a disruption of ccpA or hprK. The tglpFK‐requiring CCR mechanism seems to be based on insufficient synthesis of glycerol‐3‐P, as CCR of glpFK was absent in ccpA mutants growing on glycerol‐3‐P or synthesizing H230R mutant GlpK. In cells growing on glycerol, glucose prevents the phosphorylation of GlpK by P~His‐HPr. P~GlpK is much more active than GlpK, and the absence of P~GlpK formation in ΔptsHI strains prevents glycerol metabolism. As a consequence, only small amounts of glycerol‐3‐P will be formed in glycerol and glucose‐exposed cells (inducer exclusion). The uptake of glycerol‐3‐P via GlpT provides high concentrations of this metabolite in the ccpA mutant and allows the expression of the glpF′–lacZ fusion even when glucose is present. Similarly, despite the presence of glucose, large amounts of glycerol‐3‐P are formed in a glycerol‐exposed strain synthesizing GlpKH230R, as this mutant GlpK is as active as P~GlpK.


Molecular Microbiology | 2003

CcpA regulation of aerobic and respiration growth in Lactococcus lactis

Philippe Gaudu; Gilles Lamberet; Sandrine Poncet; Alexandra Gruss

The catabolic control protein CcpA is the highly conserved regulator of carbon metabolism in Gram‐positive bacteria. We recently showed that Lactococcus lactis, a fermenting bacterium in the family of Streptococcaceae, is capable of respiration late in growth when haem is added to aerated cultures. As the start of respiration coincides with glucose depletion from the medium, we hypothesized that CcpA is involved in this metabolic switch and investigated its role in lactococcal growth under aeration and respiration conditions. Compared with modest changes observed in fermentation growth, inactivation of ccpA shifts metabolism to mixed acid fermentation under aeration conditions. This shift is due to a modification of the redox balance via derepression of NADH oxidase, which eliminates oxygen and decreases the NADH pool. CcpA also plays a decisive role in respiration metabolism. Haem addition to lag phase ccpA cells results in growth arrest and cell mortality. Toxicity is due to oxidative stress provoked by precocious haem uptake. We identify the repressor of the haem transport system and show that it is a target of CcpA activation. We propose that CcpA‐mediated repression of haem uptake is a means of preventing oxidative damage at the start of exponential growth. CcpA thus appears to govern a regulatory network that coordinates oxygen, iron and carbon metabolism.


Proceedings of the National Academy of Sciences of the United States of America | 2002

X-ray structure of a bifunctional protein kinase in complex with its protein substrate HPr

Sonia Fieulaine; Solange Moréra; Sandrine Poncet; Ivan Mijakovic; Anne Galinier; Joël Janin; Josef Deutscher; Sylvie Nessler

HPr kinase/phosphorylase (HprK/P) controls the phosphorylation state of the phosphocarrier protein HPr and regulates the utilization of carbon sources by Gram-positive bacteria. It catalyzes both the ATP-dependent phosphorylation of Ser-46 of HPr and its dephosphorylation by phosphorolysis. The latter reaction uses inorganic phosphate as substrate and produces pyrophosphate. We present here two crystal structures of a complex of the catalytic domain of Lactobacillus casei HprK/P with Bacillus subtilis HPr, both at 2.8-Å resolution. One of the structures was obtained in the presence of excess pyrophosphate, reversing the phosphorolysis reaction and contains serine-phosphorylated HPr. The complex has six HPr molecules bound to the hexameric kinase. Two adjacent enzyme subunits are in contact with each HPr molecule, one through its active site and the other through its C-terminal helix. In the complex with serine-phosphorylated HPr, a phosphate ion is in a position to perform a nucleophilic attack on the phosphoserine. Although the mechanism of the phosphorylation reaction resembles that of eukaryotic protein kinases, the dephosphorylation by inorganic phosphate is unique to the HprK/P family of kinases. This study provides the structure of a protein kinase in complex with its protein substrate, giving insights into the chemistry of the phospho-transfer reactions in both directions.


Journal of Molecular Microbiology and Biotechnology | 2003

Transcription Regulators Potentially Controlled by HPr Kinase/Phosphorylase in Gram-Negative Bacteria

Grégory Boël; Ivan Mijakovic; Alain Mazé; Sandrine Poncet; Muhamed-Kheir Taha; Mireille Larribe; Emmanuelle Darbon; Arbia Khemiri; Anne Galinier; Josef Deutscher

Phosphorylation and dephosphorylation at Ser-46 in HPr, a phosphocarrier protein of the phosphoenolpyruvate:carbohydrate phosphotransferase system (PTS) is controlled by the bifunctional HPr kinase/phosphorylase (HprK/P). In Gram-positive bacteria, P-Ser-HPr controls (1) sugar uptake via the PTS; (2) catabolite control protein A (CcpA)-mediated carbon catabolite repression, and (3) inducer exclusion. Genome sequencing revealed that HprK/P is absent from Gram-negative enteric bacteria, but present in many other proteobacteria. These organisms also possess (1) HPr, the substrate for HprK/P; (2) enzyme I, which phosphorylates HPr at His-15, and (3) one or several enzymes IIA, which receive the phosphoryl group from P∼His-HPr. The genes encoding the PTS proteins are often organized in an operon with hprK. However, most of these organisms miss CcpA and a functional PTS, as enzymes IIB and membrane-integrated enzymes IIC seem to be absent. HprK/P and the rudimentary PTS phosphorylation cascade in Gram-negative bacteria must therefore carry out functions different from those in Gram-positive organisms. The gene organization in many HprK/P-containing Gram-negative bacteria as well as some preliminary experiments suggest that HprK/P might control transcription regulators implicated in cell adhesion and virulence. In α-proteobacteria, hprK is located downstream of genes encoding a two-component system of the EnvZ/OmpR family. In several other proteobacteria, hprK is organized in an operon together with genes from the rpoN region of Escherichia coli (rpoN encodes a σ54). We propose that HprK/P might control the phosphorylation state of HPr and EIIAs, which in turn could control the transcription regulators.


Journal of Molecular Microbiology and Biotechnology | 2005

How Seryl-Phosphorylated HPr Inhibits PrfA, a Transcription Activator of Listeria monocytogenes Virulence Genes

Rana Herro; Sandrine Poncet; Pascale Cossart; Carmen Buchrieser; Edith Gouin; Philippe Glaser; Josef Deutscher

Listeria monocytogenes PrfA, a transcription activator for several virulence genes, including the hemolysin-encoding hly, is inhibited by rapidly metabolizable carbon sources (glucose, fructose, etc.). This inhibition is not mediated via the major carbon catabolite repression mechanism of gram-positive bacteria, since inactivation of the catabolite control protein A (CcpA) did not prevent the repression of virulence genes by the above sugars. In order to test whether the catabolite co-repressor P-Ser-HPr might be involved in PrfA regulation, we used a Bacillus subtilis strain (BUG1199) containing L. monocytogenes prfA under control of pspac and the lacZ reporter gene fused to the PrfA-activated hly promoter. Formation of P-Ser-HPr requires the bifunctional HPr kinase/phosphorylase (HprK/P), which, depending on the concentration of certain metabolites, either phosphorylates HPr at Ser-46 or dephosphorylates P-Ser-HPr. The hprKV267F allele codes for an HprK/P leading to the accumulation of P-Ser-HPr, since it has normal kinase, but almost no phosphorylase activity. Interestingly, introducing hprKV267F into BUG1199 strongly inhibited transcription activation by PrfA. Preventing the accumulation of P-Ser-HPr in the hprKV267F mutant by replacing Ser-46 in HPr with an alanine restored PrfA activity, while ccpA inactivation had no effect. Interestingly, disruption of ccpA in the hprK wild-type strain BUG1199 also led to inhibition of PrfA. The lowered lacZ expression in the ccpA strain is probably also due to elevated amounts of P-Ser-HPr, since it disappeared when Ser-46 in HPr was replaced with an alanine. To carry out its catalytic function in sugar transport, HPr of the phosphotransferase system (PTS) is also phosphorylated by phosphoenolpyruvate and enzyme I at His-15. However, P-Ser-HPr is only very slowly phosphorylated by enzyme I, which probably accounts for PrfA inhibition. In agreement with this concept, disruption of the enzyme I- or HPr-encoding genes also strongly inhibited PrfA activity. PrfA activity therefore seems to depend on a fully functional PTS phosphorylation cascade.

Collaboration


Dive into the Sandrine Poncet's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sylvie Nessler

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alain Mazé

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Anne Galinier

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Grégory Boël

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Ivan Mijakovic

Chalmers University of Technology

View shared research outputs
Top Co-Authors

Avatar

Ivan Mijakovic

Chalmers University of Technology

View shared research outputs
Top Co-Authors

Avatar

Sonia Fieulaine

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge