Network


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

Hotspot


Dive into the research topics where Cédric Badiou is active.

Publication


Featured researches published by Cédric Badiou.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Polymorphonuclear leukocytes mediate Staphylococcus aureus Panton-Valentine leukocidin-induced lung inflammation and injury

Binh An Diep; Liana Chan; Pierre Tattevin; Osamu Kajikawa; Thomas R. Martin; Li Basuino; Thuy T. Mai; Helene Marbach; Kevin R. Braughton; Adeline R. Whitney; Donald J. Gardner; Xuemo Fan; Ching W. Tseng; George Y. Liu; Cédric Badiou; Jerome Etienne; Gerard Lina; Michael A. Matthay; Frank R. DeLeo; Henry F. Chambers

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) is epidemic in the United States, even rivaling HIV/AIDS in its public health impact. The pandemic clone USA300, like other CA-MRSA strains, expresses Panton-Valentine leukocidin (PVL), a pore-forming toxin that targets polymorphonuclear leukocytes (PMNs). PVL is thought to play a key role in the pathogenesis of necrotizing pneumonia, but data from rodent infection models are inconclusive. Rodent PMNs are less susceptible than human PMNs to PVL-induced cytolysis, whereas rabbit PMNs, like those of humans, are highly susceptible to PVL-induced cytolysis. This difference in target cell susceptibility could affect results of experimental models. Therefore, we developed a rabbit model of necrotizing pneumonia to compare the virulence of a USA300 wild-type strain with that of isogenic PVL-deletion mutant and -complemented strains. PVL enhanced the capacity of USA300 to cause severe lung necrosis, pulmonary edema, alveolar hemorrhage, hemoptysis, and death, hallmark clinical features of fatal human necrotizing pneumonia. Purified PVL instilled directly into the lung caused lung inflammation and injury by recruiting and lysing PMNs, which damage the lung by releasing cytotoxic granule contents. These findings provide insights into the mechanism of PVL-induced lung injury and inflammation and demonstrate the utility of the rabbit for studying PVL-mediated pathogenesis.


Antimicrobial Agents and Chemotherapy | 2007

Effect of antibiotics on Staphylococcus aureus producing Panton-Valentine leukocidin.

Oana Dumitrescu; Sandrine Boisset; Cédric Badiou; Michèle Bes; Yvonne Benito; Marie-Elisabeth Reverdy; François Vandenesch; Jerome Etienne; Gerard Lina

ABSTRACT We examined the capacity of Staphylococcus aureus strains to release Panton-Valentine leukocidin (PVL) in the presence of antibiotics. No PVL was detected when S. aureus was incubated at inhibitory concentrations, while subinhibitory concentrations of oxacillin enhanced the PVL level; clindamycin, linezolid, and fusidic acid were inhibitory; and vancomycin had roughly no effect.


Clinical Microbiology and Infection | 2009

The Panton-Valentine leukocidin vaccine protects mice against lung and skin infections caused by Staphylococcus aureus USA300.

Eric L. Brown; Oana Dumitrescu; Damien Thomas; Cédric Badiou; E.M. Koers; P. Choudhury; Vanessa Vazquez; J. Etienne; Gerard Lina; François Vandenesch; Maria Gabriela Bowden

Methicillin-resistant Staphylococcus aureus is increasingly responsible for staphylococcal infections in the community. A large percentage of the community-acquired methicillin-resistant (CA-MRSA) strains in the USA produce Panton-Valentine leukocidin (PVL), which is associated with severe infections. The virulence of the clinical CA-MRSA strain USA300 was compared to that of its isogenic pvl-deleted mutant, and it was shown that PVL contributes to lung and muscle tissue destruction, respectively, in murine necrotizing pneumonia and skin infection models. Mice infected with the USA300 strain developed a dominant anti-PVL response. The PVL subunits were therefore tested as vaccinogens against this isolate, and their vaccine efficacy correlated with both the route of vaccination and infection. These data suggest that PVL is a virulence factor in murine CA-MRSA infections.


Cell Host & Microbe | 2013

The Staphylococcal Toxin Panton-Valentine Leukocidin Targets Human C5a Receptors

András N. Spaan; Thomas Henry; Willemien J. M. van Rooijen; Magali Perret; Cédric Badiou; Piet C. Aerts; Johan Kemmink; Carla J. C. de Haas; Kok P. M. van Kessel; François Vandenesch; Gerard Lina; Jos A. G. van Strijp

Panton-Valentine Leukocidin (PVL) is a staphylococcal bicomponent pore-forming toxin linked to severe invasive infections. Target-cell and species specificity of PVL are poorly understood, and the mechanism of action of this toxin in Staphylococcus aureus virulence is controversial. Here, we identify the human complement receptors C5aR and C5L2 as host targets of PVL, mediating both toxin binding and cytotoxicity. Expression and interspecies variations of the C5aR determine cell and species specificity of PVL. The C5aR binding PVL component, LukS-PV, is a potent inhibitor of C5a-induced immune cell activation. These findings provide insight into leukocidin function and staphylococcal virulence and offer directions for future investigations into individual susceptibility to severe staphylococcal disease.


Clinical Microbiology and Infection | 2008

Effect of antibiotics, alone and in combination, on Panton-Valentine leukocidin production by a Staphylococcus aureus reference strain.

Oana Dumitrescu; Cédric Badiou; Michèle Bes; Marie Elisabeth Reverdy; François Vandenesch; Jerome Etienne; Gerard Lina

The capacity of Staphylococcus aureus strain LUG855 to release Panton-Valentine leukocidin (PVL) in the presence of sub-inhibitory concentrations of anti-staphylococcal drugs was examined. Oxacillin enhanced PVL release 2.5-fold, while clindamycin, linezolid, fusidic acid and rifampicin were inhibitory, and vancomycin, pristinamycin, tetracycline, ofloxacin and co-trimoxazole had no effect. In combination with oxacillin, sub-inhibitory concentrations of clindamycin or rifampicin inhibited PVL induction significantly, linezolid was less inhibitory, and fusidic acid did not inhibit PVL induction by oxacillin. These data support the use of oxacillin in combination with clindamycin, rifampicin or linezolid for the treatment of PVL-positive S. aureus infections.


PLOS ONE | 2011

Panton-Valentine Leukocidin Does Play a Role in the Early Stage of Staphylococcus aureus Skin Infections: A Rabbit Model

Urszula Lipinska; Katleen Hermans; Lieve Meulemans; Oana Dumitrescu; Cédric Badiou; Luc Duchateau; Freddy Haesebrouck; Jerome Etienne; Gerard Lina

Despite epidemiological data linking necrotizing skin infections with the production of Panton-Valentine leukocidin (PVL), the contribution of this toxin to the virulence of S. aureus has been highly discussed as a result of inconclusive results of in vivo studies. However, the majority of these results originate from experiments using mice, an animal species which neutrophils - the major target cells for PVL - are highly insensitive to the action of this leukocidin. In contrast, the rabbit neutrophils have been shown to be as sensitive to PVL action as human cells, making the rabbit a better experimental animal to explore the PVL role. In this study we examined whether PVL contributes to S. aureus pathogenicity by means of a rabbit skin infection model. The rabbits were injected intradermally with 108 cfu of either a PVL positive community-associated methicillin-resistant S. aureus isolate, its isogenic PVL knockout or a PVL complemented knockout strain, and the development of skin lesions was observed. While all strains induced skin infection, the wild type strain produced larger lesions and a higher degree of skin necrosis compared to the PVL knockout strain in the first week after the infection. The PVL expression in the rabbits was indirectly confirmed by a raise in the serum titer of anti-LukS-PV antibodies observed only in the rabbits infected with PVL positive strains. These results indicate that the rabbit model is more suitable for studying the role of PVL in staphylococcal diseases than other animal models. Further, they support the epidemiological link between PVL producing S. aureus strains and necrotizing skin infections.


Clinical Infectious Diseases | 2010

Immunogenicity of Toxins during Staphylococcus aureus Infection

Nelianne J. Verkaik; Olivier Dauwalder; Kenza Antri; Ilhem Boubekri; Corné P. de Vogel; Cédric Badiou; Michèle Bes; Francçois Vandenesch; Mohamed Tazir; Herbert Hooijkaas; Henri A. Verbrugh; Alex van Belkum; Jerome Etienne; Gerard Lina; Nadjia Ramdani-Bouguessa; Willem J. B. van Wamel

BACKGROUND Toxins are important Staphylococcus aureus virulence factors, but little is known about their immunogenicity during infection. Here, additional insight is generated. METHODS Serum samples from 206 S. aureus-infected patients and 201 hospital-admitted control subjects were analyzed for immunoglobulin (Ig) G binding to 20 toxins, using flow-cytometry based technology. Antibody levels were associated with polymerase chain reaction-defined presence of toxin genes in homologous S. aureus isolates. RESULTS IgG levels directed to exfoliative toxin (ET) A, ETB, gamma hemolysin B (HlgB), leukocidin (Luk) D, LukE, LukS, staphylococcal enterotoxin (SE) A, SEE, SEH, SEI, and SElM were higher in S. aureus-infected patients than in control subjects (P < .05). Furthermore, in the S. aureus-infected patient group, IgG levels were higher if genes encoding ETA, ETB, SEA, SEC, SEH, SElQ, toxic shock syndrome toxin-1 (TSST-1), or Panton-Valentine leukocidin (PVL) were present in the infectious isolate (P< .05). Levels of anti-SEA IgG increased during infections with sea-positive (median fluorescence intensity from 11,555 to 12,388; P<.05) but not sea-negative strains. In addition, anti-LukS IgG levels increased during skin and soft-tissue infections with luk-PV-positive (median fluorescence intensity from 15,231 to 15,911; P<.05) but not luk-PV-negative strains. Bacteremia was associated with sea (odds ratio, 3.4; 95% confidence interval, 1.2-10.0) and tst (odds ratio, 5.7; 95% confidence interval, 1.6-20.8). Skin and soft-tissue infections and bone and joint infections were associated with luk-PV (odds ratio, 2.5; 95% confidence interval, 1.2-5.2). CONCLUSIONS Many toxins are expressed in vivo and recognized by the immune system during staphylococcal infections, suggesting their involvement in S. aureus pathogenesis.


Nature Communications | 2014

The staphylococcal toxins γ-haemolysin AB and CB differentially target phagocytes by employing specific chemokine receptors

András N. Spaan; Manouk Vrieling; Pierre Wallet; Cédric Badiou; Tamara Reyes-Robles; Elizabeth A. Ohneck; Yvonne Benito; Carla J. C. de Haas; Christopher P. Day; Michael P. Jennings; Gerard Lina; François Vandenesch; Kok P. M. van Kessel; Victor J. Torres; Jos A. G. van Strijp; Thomas Henry

Evasion of the host phagocyte response by Staphylococcus aureus is crucial to successful infection with the pathogen. γ-Hemolysin AB and CB (HlgAB, HlgCB) are bicomponent pore-forming toxins present in almost all human S. aureus isolates. Cellular tropism and contribution of the toxins to S. aureus pathophysiology are poorly understood. Here, we identify the chemokine receptors CXCR1, CXCR2 and CCR2 as targets for HlgAB, and the complement receptors C5aR and C5L2 as targets for HlgCB. The receptor expression patterns allow the toxins to efficiently and differentially target phagocytic cells. Murine neutrophils are resistant to HlgAB and HlgCB. CCR2 is the sole murine receptor orthologue compatible with γ-Hemolysin. In a murine peritonitis model, HlgAB contributes to S. aureus bacteremia in a CCR2-dependent manner. HlgAB-mediated targeting of CCR2+ cells highlights the involvement of inflammatory macrophages during S. aureus infection. Functional quantification identifies HlgAB and HlgCB as major secreted staphylococcal leukocidins.


Cellular Microbiology | 2012

Cross‐talk between Staphylococcus aureus leukocidins‐intoxicated macrophages and lung epithelial cells triggers chemokine secretion in an inflammasome‐dependent manner

Magali Perret; Cédric Badiou; Gerard Lina; Sophie Burbaud; Yvonne Benito; Michèle Bes; Vincent Cottin; Florence Couzon; Carole Juruj; Olivier Dauwalder; Nadège Goutagny; Binh An Diep; François Vandenesch; Thomas Henry

Staphylococcus aureus is a major pathogen responsible for both nosocomial and community‐acquired infections. Central to its virulence is its ability to secrete haemolysins, pore‐forming toxins and cytolytic peptides. The large number of membrane‐damaging toxins and peptides produced during S. aureus infections has hindered a precise understanding of their specific roles in diseases. Here, we used comprehensive libraries of recombinant toxins and synthetic cytolytic peptides, of S. aureus mutants and clinical strains to investigate the role of these virulence factors in targeting human macrophages and triggering IL‐1β release. We found that the Panton Valentine leukocidin (PVL) is the major trigger of IL‐1β release and inflammasome activation in primary human macrophages. The cytolytic peptides, δ‐haemolysin and PSMα3; the pore‐forming toxins, γ‐haemolysin and LukDE; and β‐haemolysin synergize with PVL to amplify IL‐1β release, indicating that these factors cooperate with PVL to trigger inflammation. PVL+S. aureus causes necrotizing pneumonia in children and young adults. The severity of this disease is due to the massive recruitment of neutrophils that cause lung damage. Importantly, we demonstrate that PVL triggers IL‐1β release in human alveolar macrophages. Furthermore, IL‐1β released by PVL‐intoxicated macrophages stimulates the secretion of the neutrophil attracting chemokines, IL‐8 and monocyte chemotactic protein‐1, by lung epithelial cells. Finally, we show that PVL‐induced IL‐8/monocyte chemotactic protein‐1 release is abolished by the inclusion of IL‐1 receptor antagonist (IL‐1Ra) in a mixed culture of lung epithelial cells and macrophages. Together, our results identify PVL as the predominant S. aureus secreted factor for triggering inflammasome activation in human macrophages and demonstrate how PVL‐intoxicated macrophages orchestrate inflammation in the lung. Finally, our work suggests that anakinra, a synthetic IL‐1Ra, may be an effective therapeutic agent to reduce the massive neutrophils infiltration observed during necrotizing pneumonia and decrease the resulting host‐mediated lung injury.


PLOS ONE | 2013

PSMs of hypervirulent Staphylococcus aureus act as intracellular toxins that kill infected osteoblasts.

Jean-Philippe Rasigade; Sophie Trouillet-Assant; Tristan Ferry; Binh An Diep; Anaı̈s Sapin; Yannick Lhoste; Jérémy Ranfaing; Cédric Badiou; Yvonne Benito; Michèle Bes; Florence Couzon; Sylvestre Tigaud; Gerard Lina; Jerome Etienne; François Vandenesch; Frédéric Laurent

Epidemic community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA) is associated with more severe and acute forms of osteomyelitis than healthcare-associated (HA-) MRSA. Although S. aureus is now recognized as a facultative intracellular pathogen, the contribution of osteoblast invasion by CA-MRSA to the pathogenesis of osteomyelitis is unknown. Using an ex vivo model of intracellular infection of human osteoblasts, we demonstrated that CA-MRSA strains of diverse lineages share an enhanced ability to kill infected osteoblasts compared to HA-MRSA. Cytotoxicity comparisons of CA-MRSA isogenic deletion mutants revealed that phenol-soluble modulins (PSMs), a class of membrane-damaging exoproteins that are expressed at higher levels in CA-MRSA than in HA-MRSA, are involved in this osteoblast killing, whereas other major CA-MRSA virulence determinants, the Panton-Valentine leukocidin and alpha-toxin, are not involved. Similarly, functional agr and sarA regulators, which control the expression of PSMs and alpha-toxin, were required for the expression of the intracellular cytotoxic phenotype by CA-MRSA, whereas the saeRS regulator, which controls the expression of alpha-toxin but not PSMs, had no impact on cytotoxicity. Finally, PSM transcript levels determined by quantitative reverse-transcriptase PCR were significantly higher in CA-MRSA than in HA-MRSA strains and associated with cell damage in MRSA-infected osteoblasts. These findings provide new insights into the pathogenesis of severe CA-MRSA osteomyelitis and unravel a novel virulence strategy of CA-MRSA, based on the invasion and subsequent killing of osteoblasts by PSMs acting as intracellular toxins.

Collaboration


Dive into the Cédric Badiou's collaboration.

Top Co-Authors

Avatar

Gerard Lina

École normale supérieure de Lyon

View shared research outputs
Top Co-Authors

Avatar

François Vandenesch

École normale supérieure de Lyon

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michèle Bes

École normale supérieure de Lyon

View shared research outputs
Top Co-Authors

Avatar

Binh An Diep

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge