16 research outputs found

    LpxC inhibitors evaluation : a new promising antimicrobial class

    No full text
    L’émergence et la diffusion de la rĂ©sistance aux antibiotiques au sein des bactĂ©ries Ă  Gram nĂ©gatif (BGN) sont aujourd’hui des enjeux de SantĂ© Publique nationaux et internationaux. La multi-rĂ©sistance aux antibiotiques concerne non seulement des espĂšces frĂ©quemment responsables d’infections nosocomiales mais aussi des espĂšces hautement virulentes comme Yersinia pestis, agent de la peste et du bioterrorisme. Dans ce contexte, la mise au point de nouvelles molĂ©cules actives sur d’autres cibles bactĂ©riennes est primordiale. La mĂ©tallo-enzyme LpxC catalyse la premiĂšre Ă©tape irrĂ©versible de la biosynthĂšse du lipide A, constituant majeur de la membrane externe des bactĂ©ries Ă  Gram nĂ©gatif. Des inhibiteurs de LpxC sont ainsi dĂ©veloppĂ©s depuis une vingtaine d’annĂ©es mais leur spectre sur les BGN Ă©tait initialement limitĂ© aux entĂ©robactĂ©ries et leur activitĂ© partielle sur P. aeruginosa. Dans ce travail nous avons participĂ© Ă  l’optimisation de la structure chimique de ces molĂ©cules grĂące Ă  une approche dynamique des interactions enzymes/inhibiteurs utilisant la rĂ©sonance magnĂ©tique nuclĂ©aire (RMN). Cette technique a permis l’élaboration d’un nouvel inhibiteur de LpxC, le LPC-058, caractĂ©risĂ© par une forte affinitĂ© pour l’enzyme (Ki = 3,5 ± 0,2 pM). Nous avons Ă©valuĂ© in vitro l’activitĂ© antibiotique du LPC-058 et de trois autres composĂ©s (CHIR-090, LPC-011 et LPC-087) vis-Ă -vis de 369 souches cliniques responsables d’infections nosocomiales aux profils de rĂ©sistance variĂ©s. Le LPC-058 prĂ©sentait le plus large spectre d’activitĂ© en particulier sur A. baumannii et les valeurs de CMI les plus basses (CMI90 = 0,12 mg/L pour les entĂ©robactĂ©ries et 0,5 mg/L pour P. aeruginosa). Il Ă©tait bactĂ©ricide vis-Ă -vis de souches multi-rĂ©sistantes et son action Ă©tait synergique avec les C3G, l’imipĂ©nĂšme, l’amikacine et la ciprofloxacine vis-Ă -vis de souches de K. pneumoniae, P. aeruginosa et A. baumannii productrice de carbapĂ©nĂ©mases, respectivement KPC-2, VIM-1 et OXA-23. Le LPC-058 prĂ©sentait nĂ©anmoins une forte fixation protĂ©ique et, in vivo, son volume de distribution Ă©tait limitĂ© au compartiment sanguin (Vd = 1,1 L/kg). Nous avons Ă©valuĂ© son activitĂ© in vivo dans un modĂšle murin de peste bubonique car il s’agit de l’une des infections les plus virulentes pour l’homme. Nous avons obtenu une survie de 87 % aprĂšs 5 jours de traitement Ă  la posologie de 10 mg/kg q8h par voie veineuse. Le LPC-058 occasionnant des diarrhĂ©es chez le rongeur, nous avons Ă©valuĂ© un de ses dĂ©rivĂ©s, le LPC-B, caractĂ©risĂ© par une moindre fixation protĂ©ique, un plus grand volume de distribution et l’absence d’effets secondaires chez la souris, mĂȘme Ă  fortes doses. Nous avons dĂ©montrĂ© qu’à la posologie de 200 mg/kg par voie veineuse, cet antibiotique Ă©tait aussi efficace que la doxycycline (traitement de rĂ©fĂ©rence de la peste). L’ensemble de ces travaux souligne le rĂŽle potentiel des inhibiteurs de LpxC dans la prise en charge des infections par des bactĂ©ries multi-rĂ©sistantes ou hautement virulentes.Antimicrobial resistance among Gram-negative bacteria (GNB) has become a national and international public health concern. Resistant strains are involved in nosocomial diseases and in highly virulent infections, such as plague caused by Yersinia pestis, a potential biological terrorism agent. In this context the development of new antimicrobial compounds efficient on new bacterial targets is critical. LpxC metallo-enzyme catalyzes the first commitment step of the lipid A biosynthesis, a major component of the Gram negative cell wall. LpxC inhibitors have been developed for twenty years but their activity was restricted to enterobacteria and weak against Pseudomonas aeruginosa. In this study, we have collaborated in the chemical optimization of the compounds thanks to a dynamic approach of enzyme/inhibitor interactions brought by nuclear magnetic resonance (NMR). This technology enabled the development of LPC-058, a new inhibitor, showing a high potency against LpxC (Ki = 3.5 ± 0.2 pM). We studied the in vitro efficacy of LPC-058 and three other compounds (CHIR-090, LPC-011 and LPC-087) against 369 clinical strains responsible for nosocomial infections with various antibiotic resistance profiles. In this part, LPC-058 displayed the broadest spectrum of efficacy, even on Acinetobacter baumannii with the lowest MIC values (MIC90 = 0.12 mg/L against enterobacteria and 0.5 mg/L against P. aeruginosa). It showed bactericidal activity against multi-resistant strains and synergistic activity in association with third generation cephalosporins, imipenem, amikacin and ciprofloxacin against carbapenemase producing Klebsiella pneumoniae, P. aeruginosa et A. baumannii strains (respectively KPC-2, VIM-1 and OXA-23). However, LPC-058 was constrained by strong protein interactions and a small volume of distribution (Vd = 1.1 L/kg). In vivo efficacy was studied in a murine model of bubonic plague. A 87% survival rate was obtained after five days of 10 mg/kg q8h intravenous administration. As LPC-058 treatment was associated to diarrheas in mice, we evaluated another derivate, LPC-B, characterized by a larger volume of distribution, minor protein fixation and less side effects, even for a high dose posology. We demonstrated a comparable efficacy between 200 mg/kg LPC-B treatment and doxycyclin administration (recommended in plague treatment). This work highlights the potential use of LpxC inhibitors in the management of infections caused by multi-resistant or highly virulent Gram-negative bacteria

    Evaluation d’une nouvelle classe d’antibiotiques : les inhibiteurs de LpxC

    No full text
    Antimicrobial resistance among Gram-negative bacteria (GNB) has become a national and international public health concern. Resistant strains are involved in nosocomial diseases and in highly virulent infections, such as plague caused by Yersinia pestis, a potential biological terrorism agent. In this context the development of new antimicrobial compounds efficient on new bacterial targets is critical. LpxC metallo-enzyme catalyzes the first commitment step of the lipid A biosynthesis, a major component of the Gram negative cell wall. LpxC inhibitors have been developed for twenty years but their activity was restricted to enterobacteria and weak against Pseudomonas aeruginosa. In this study, we have collaborated in the chemical optimization of the compounds thanks to a dynamic approach of enzyme/inhibitor interactions brought by nuclear magnetic resonance (NMR). This technology enabled the development of LPC-058, a new inhibitor, showing a high potency against LpxC (Ki = 3.5 ± 0.2 pM). We studied the in vitro efficacy of LPC-058 and three other compounds (CHIR-090, LPC-011 and LPC-087) against 369 clinical strains responsible for nosocomial infections with various antibiotic resistance profiles. In this part, LPC-058 displayed the broadest spectrum of efficacy, even on Acinetobacter baumannii with the lowest MIC values (MIC90 = 0.12 mg/L against enterobacteria and 0.5 mg/L against P. aeruginosa). It showed bactericidal activity against multi-resistant strains and synergistic activity in association with third generation cephalosporins, imipenem, amikacin and ciprofloxacin against carbapenemase producing Klebsiella pneumoniae, P. aeruginosa et A. baumannii strains (respectively KPC-2, VIM-1 and OXA-23). However, LPC-058 was constrained by strong protein interactions and a small volume of distribution (Vd = 1.1 L/kg). In vivo efficacy was studied in a murine model of bubonic plague. A 87% survival rate was obtained after five days of 10 mg/kg q8h intravenous administration. As LPC-058 treatment was associated to diarrheas in mice, we evaluated another derivate, LPC-B, characterized by a larger volume of distribution, minor protein fixation and less side effects, even for a high dose posology. We demonstrated a comparable efficacy between 200 mg/kg LPC-B treatment and doxycyclin administration (recommended in plague treatment). This work highlights the potential use of LpxC inhibitors in the management of infections caused by multi-resistant or highly virulent Gram-negative bacteria.L’émergence et la diffusion de la rĂ©sistance aux antibiotiques au sein des bactĂ©ries Ă  Gram nĂ©gatif (BGN) sont aujourd’hui des enjeux de SantĂ© Publique nationaux et internationaux. La multi-rĂ©sistance aux antibiotiques concerne non seulement des espĂšces frĂ©quemment responsables d’infections nosocomiales mais aussi des espĂšces hautement virulentes comme Yersinia pestis, agent de la peste et du bioterrorisme. Dans ce contexte, la mise au point de nouvelles molĂ©cules actives sur d’autres cibles bactĂ©riennes est primordiale. La mĂ©tallo-enzyme LpxC catalyse la premiĂšre Ă©tape irrĂ©versible de la biosynthĂšse du lipide A, constituant majeur de la membrane externe des bactĂ©ries Ă  Gram nĂ©gatif. Des inhibiteurs de LpxC sont ainsi dĂ©veloppĂ©s depuis une vingtaine d’annĂ©es mais leur spectre sur les BGN Ă©tait initialement limitĂ© aux entĂ©robactĂ©ries et leur activitĂ© partielle sur P. aeruginosa. Dans ce travail nous avons participĂ© Ă  l’optimisation de la structure chimique de ces molĂ©cules grĂące Ă  une approche dynamique des interactions enzymes/inhibiteurs utilisant la rĂ©sonance magnĂ©tique nuclĂ©aire (RMN). Cette technique a permis l’élaboration d’un nouvel inhibiteur de LpxC, le LPC-058, caractĂ©risĂ© par une forte affinitĂ© pour l’enzyme (Ki = 3,5 ± 0,2 pM). Nous avons Ă©valuĂ© in vitro l’activitĂ© antibiotique du LPC-058 et de trois autres composĂ©s (CHIR-090, LPC-011 et LPC-087) vis-Ă -vis de 369 souches cliniques responsables d’infections nosocomiales aux profils de rĂ©sistance variĂ©s. Le LPC-058 prĂ©sentait le plus large spectre d’activitĂ© en particulier sur A. baumannii et les valeurs de CMI les plus basses (CMI90 = 0,12 mg/L pour les entĂ©robactĂ©ries et 0,5 mg/L pour P. aeruginosa). Il Ă©tait bactĂ©ricide vis-Ă -vis de souches multi-rĂ©sistantes et son action Ă©tait synergique avec les C3G, l’imipĂ©nĂšme, l’amikacine et la ciprofloxacine vis-Ă -vis de souches de K. pneumoniae, P. aeruginosa et A. baumannii productrice de carbapĂ©nĂ©mases, respectivement KPC-2, VIM-1 et OXA-23. Le LPC-058 prĂ©sentait nĂ©anmoins une forte fixation protĂ©ique et, in vivo, son volume de distribution Ă©tait limitĂ© au compartiment sanguin (Vd = 1,1 L/kg). Nous avons Ă©valuĂ© son activitĂ© in vivo dans un modĂšle murin de peste bubonique car il s’agit de l’une des infections les plus virulentes pour l’homme. Nous avons obtenu une survie de 87 % aprĂšs 5 jours de traitement Ă  la posologie de 10 mg/kg q8h par voie veineuse. Le LPC-058 occasionnant des diarrhĂ©es chez le rongeur, nous avons Ă©valuĂ© un de ses dĂ©rivĂ©s, le LPC-B, caractĂ©risĂ© par une moindre fixation protĂ©ique, un plus grand volume de distribution et l’absence d’effets secondaires chez la souris, mĂȘme Ă  fortes doses. Nous avons dĂ©montrĂ© qu’à la posologie de 200 mg/kg par voie veineuse, cet antibiotique Ă©tait aussi efficace que la doxycycline (traitement de rĂ©fĂ©rence de la peste). L’ensemble de ces travaux souligne le rĂŽle potentiel des inhibiteurs de LpxC dans la prise en charge des infections par des bactĂ©ries multi-rĂ©sistantes ou hautement virulentes

    Physicochemical and Microbiological Stability of a New Oral Clonidine Solution for Paediatric Use

    No full text
    As many drugs are unavailable for paediatric use, hospital pharmacies are often required to develop suitable formulations themselves. Clonidine is commonly used in paediatrics (in severe hypertension, in opiate withdrawal syndrome, in tics and Gilles de la Tourette syndrome or in anaesthetic premedication) but no appropriate formulation has been drawn up. The aims of this work were to develop an oral solution of clonidine dedicated to children and to assess its physicochemical and microbiological stability

    Characterization of the protective immune response to Yersinia pseudotuberculosis infection in mice vaccinated with an LcrV-secreting strain of Lactococcus lactis

    No full text
    International audienceBACKGROUND: Pseudotuberculosis is an infection caused by the bacterial enteropathogen Yersinia pseudotuberculosis and is considered to be a significant problem in veterinary medicine. We previously found that intranasal administration of a recombinant Lactococcus lactis strain that secretes the low-calcium response V (LcrV) antigen from Y. pseudotuberculosis (Ll-LcrV) confers protection against a lethal Y. pseudotuberculosis infection. Here, we aimed at characterizing the immunological basis of this LcrV-elicited protective response and at determining the duration of vaccine-induced immunity.METHODS: Splenocytes from BALB/c mice intranasally immunized with Ll-LcrV or Ll as control were immunostained then analyzed by flow cytometry. Protection against a lethal intravenous injection of Y. pseudotuberculosis was also determined (i) in immunized BALB/c mice depleted or not of CD4+, CD8+ or CD25+ cells and (ii) in naĂŻve BALB/c mice receiving serum from immunized mice by counting the number of bacteria in liver and spleen. Lastly, survival rate of immunized BALB/c mice following a lethal intravenous injection of Y. pseudotuberculosis was followed up to 9-months.RESULTS: We found that T and B lymphocytes but not non-conventional lymphoid cells were affected by Ll-LcrV immunization. We also observed that depletion of CD4+ and CD25+ but not CD8+ cells in immunized mice eradicated protection against a lethal systemic Y. pseudotuberculosis infection, suggesting that activated CD4+ T lymphocytes are required for vaccine-induced protection. Adoptive transfer of LcrV-specific antibodies from Ll-LcrV-immunized animals significantly reduced the bacterial counts in the liver compared to non-vaccinated mice. Lastly, the protective immunity conferred by Ll-LcrV decreased slightly over time; nevertheless almost 60% of the mice survived a lethal bacterial challenge at 9months post-vaccination.CONCLUSION: Mucosal vaccination of mice with Ll-LcrV induced cell- and antibody-mediated protective immunity against Y. pseudotuberculosis infection in the mouse and the protection is long-lasting

    Characterization of the protective immune response to Yersinia pseudotuberculosis infection in mice vaccinated with an LcrV-secreting strain of Lactococcus lactis

    No full text
    International audienceBACKGROUND: Pseudotuberculosis is an infection caused by the bacterial enteropathogen Yersinia pseudotuberculosis and is considered to be a significant problem in veterinary medicine. We previously found that intranasal administration of a recombinant Lactococcus lactis strain that secretes the low-calcium response V (LcrV) antigen from Y. pseudotuberculosis (Ll-LcrV) confers protection against a lethal Y. pseudotuberculosis infection. Here, we aimed at characterizing the immunological basis of this LcrV-elicited protective response and at determining the duration of vaccine-induced immunity.METHODS: Splenocytes from BALB/c mice intranasally immunized with Ll-LcrV or Ll as control were immunostained then analyzed by flow cytometry. Protection against a lethal intravenous injection of Y. pseudotuberculosis was also determined (i) in immunized BALB/c mice depleted or not of CD4+, CD8+ or CD25+ cells and (ii) in naĂŻve BALB/c mice receiving serum from immunized mice by counting the number of bacteria in liver and spleen. Lastly, survival rate of immunized BALB/c mice following a lethal intravenous injection of Y. pseudotuberculosis was followed up to 9-months.RESULTS: We found that T and B lymphocytes but not non-conventional lymphoid cells were affected by Ll-LcrV immunization. We also observed that depletion of CD4+ and CD25+ but not CD8+ cells in immunized mice eradicated protection against a lethal systemic Y. pseudotuberculosis infection, suggesting that activated CD4+ T lymphocytes are required for vaccine-induced protection. Adoptive transfer of LcrV-specific antibodies from Ll-LcrV-immunized animals significantly reduced the bacterial counts in the liver compared to non-vaccinated mice. Lastly, the protective immunity conferred by Ll-LcrV decreased slightly over time; nevertheless almost 60% of the mice survived a lethal bacterial challenge at 9months post-vaccination.CONCLUSION: Mucosal vaccination of mice with Ll-LcrV induced cell- and antibody-mediated protective immunity against Y. pseudotuberculosis infection in the mouse and the protection is long-lasting

    Clinical relevance of Clostridium bacteremia: An 8-year retrospective study

    No full text
    International audienceClostridium spp. are recovered from 25% of the blood culture positive with anaerobes. However, the clinical relevance of Clostridium bacteremia has been controverted in the literature, particularly for C. perfringens. We aimed to evaluate the clinical relevance of Clostridium bacteremia, either due to C. perfringens or other Clostridium species, and to identify the risk factors of mortality in these patients. A retrospective cohort study was conducted from January 2010 to April 2018. All the patients with at least one blood culture positive with any Clostridium species were included. Eighty-one patients with a least one blood culture positive with any Clostridium species were included. Seventy patients (86.4%) fulfilled the criteria for clinically relevant bacteremia. Bacteremia due to C. perfringens tended to be less clinically relevant than other Clostridium species but this was not statistically significant (76% vs 91.2%, P = 0.09). In case of clinically relevant bacteremia, the 30-day mortality rate was 31.4%. In multivariate analysis, adequate empiric antimicrobial therapy was significantly associated with survival (P = 0.03). In conclusion, bacteremia due to C. perfringens or other Clostridium species is usually clinically relevant. This finding was also supported by an improved survival at 30 days when adequate empiric antimicrobial therapy was administered

    Distinct Immune Response in Two MERS-CoV-Infected Patients: Can We Go from Bench to Bedside?

    No full text
    International audienceOne year after the occurrence of the first case of infection by the Middle East Respiratory Syndrome coronavirus (MERS-CoV) there is no clear consensus on the best treatment to propose. The World Health Organization, as well as several other national agencies, are still working on different clinical approaches to implement the most relevant treatment in MERS-CoV infection. We compared innate and adaptive immune responses of two patients infected with MERS-CoV to understand the underlying mechanisms involved in the response and propose potential therapeutic approaches. Broncho-alveolar lavage (BAL) of the first week and sera of the first month from the two patients were used in this study. Quantitative polymerase chain reaction (qRTPCR) was performed after extraction of RNA from BAL cells of MERS-CoV infected patients and control patients. BAL supernatants and sera were used to assess cytokines and chemokines secretion by enzyme-linked immunosorbent assay. The first patient died rapidly after 3 weeks in the intensive care unit, the second patient still recovers from infection. The patient with a poor outcome (patient 1), compared to patient 2, did not promote type-1 Interferon (IFN), and particularly IFNα, in response to double stranded RNA (dsRNA) from MERS-CoV. The absence of IFNα, known to promote antigen presentation in response to viruses, impairs the development of a robust antiviral adaptive Th-1 immune response. This response is mediated by IL-12 and IFNγ that decreases viral clearance; levels of both of these mediators were decreased in patient 1. Finally, we confirm previous in vitro findings that MERS-CoV can drive IL-17 production in humans. Host recognition of viral dsRNA determines outcome in the early stage of MERS-CoV infection. We highlight the critical role of IFNα in this initial stage to orchestrate a robust immune response and bring substantial arguments for the indication of early IFNα treatment during MERS-CoV infection

    Chromosome folding and prophage activation reveal specific genomic architecture for intestinal bacteria

    No full text
    International audienceBackground Bacteria and their viruses, bacteriophages, are the most abundant entities of the gut microbiota, a complex community of microorganisms associated with human health and disease. In this ecosystem, the interactions between these two key components are still largely unknown. In particular, the impact of the gut environment on bacteria and their associated prophages is yet to be deciphered. Results To gain insight into the activity of lysogenic bacteriophages within the context of their host genomes, we performed proximity ligation-based sequencing (Hi-C) in both in vitro and in vivo conditions on the 12 bacterial strains of the OMM 12 synthetic bacterial community stably associated within mice gut (gnotobiotic mouse line OMM 12 ). High-resolution contact maps of the chromosome 3D organization of the bacterial genomes revealed a wide diversity of architectures, differences between environments, and an overall stability over time in the gut of mice. The DNA contacts pointed at 3D signatures of prophages leading to 16 of them being predicted as functional. We also identified circularization signals and observed different 3D patterns between in vitro and in vivo conditions. Concurrent virome analysis showed that 11 of these prophages produced viral particles and that OMM 12 mice do not carry other intestinal viruses. Conclusions The precise identification by Hi-C of functional and active prophages within bacterial communities will unlock the study of interactions between bacteriophages and bacteria across conditions (healthy vs disease)
    corecore