Network


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

Hotspot


Dive into the research topics where Daniel Ritz is active.

Publication


Featured researches published by Daniel Ritz.


Molecular Microbiology | 2009

Second messenger signalling governs Escherichia coli biofilm induction upon ribosomal stress.

Alex Boehm; Samuel Steiner; Franziska Zaehringer; Alain Casanova; Fabienne Hamburger; Daniel Ritz; Wolfgang Keck; Martin Ackermann; Tilman Schirmer; Urs Jenal

Biofilms are communities of surface‐attached, matrix‐embedded microbial cells that can resist antimicrobial chemotherapy and contribute to persistent infections. Using an Escherichia coli biofilm model we found that exposure of bacteria to subinhibitory concentrations of ribosome‐targeting antibiotics leads to strong biofilm induction. We present evidence that this effect is elicited by the ribosome in response to translational stress. Biofilm induction involves upregulation of the polysaccharide adhesin poly‐β‐1,6‐N‐acetyl‐glucosamine (poly‐GlcNAc) and two components of the poly‐GlcNAc biosynthesis machinery, PgaA and PgaD. Poly‐GlcNAc control depends on the bacterial signalling molecules guanosine‐bis 3′, 5′(diphosphate) (ppGpp) and bis‐(3′‐5′)‐cyclic di‐GMP (c‐di‐GMP). Treatment with translation inhibitors causes a ppGpp hydrolase (SpoT)‐mediated reduction of ppGpp levels, resulting in specific derepression of PgaA. Maximal induction of PgaD and poly‐GlcNAc synthesis requires the production of c‐di‐GMP by the dedicated diguanylate cyclase YdeH. Our results identify a novel regulatory mechanism that relies on ppGpp signalling to relay information about ribosomal performance to the Pga machinery, thereby inducing adhesin production and biofilm formation. Based on the important synergistic roles of ppGpp and c‐di‐GMP in this process, we suggest that interference with bacterial second messenger signalling might represent an effective means for biofilm control during chronic infections.


PLOS Pathogens | 2010

YfiBNR Mediates Cyclic di-GMP Dependent Small Colony Variant Formation and Persistence in Pseudomonas aeruginosa

Jacob G. Malone; Tina Jaeger; Christian Spangler; Daniel Ritz; Anne Spang; Cécile Arrieumerlou; Regine Landmann; Urs Jenal

During long-term cystic fibrosis lung infections, Pseudomonas aeruginosa undergoes genetic adaptation resulting in progressively increased persistence and the generation of adaptive colony morphotypes. This includes small colony variants (SCVs), auto-aggregative, hyper-adherent cells whose appearance correlates with poor lung function and persistence of infection. The SCV morphotype is strongly linked to elevated levels of cyclic-di-GMP, a ubiquitous bacterial second messenger that regulates the transition between motile and sessile, cooperative lifestyles. A genetic screen in PA01 for SCV-related loci identified the yfiBNR operon, encoding a tripartite signaling module that regulates c-di-GMP levels in P. aeruginosa. Subsequent analysis determined that YfiN is a membrane-integral diguanylate cyclase whose activity is tightly controlled by YfiR, a small periplasmic protein, and the OmpA/Pal-like outer-membrane lipoprotein YfiB. Exopolysaccharide synthesis was identified as the principal downstream target for YfiBNR, with increased production of Pel and Psl exopolysaccharides responsible for many characteristic SCV behaviors. An yfi-dependent SCV was isolated from the sputum of a CF patient. Consequently, the effect of the SCV morphology on persistence of infection was analyzed in vitro and in vivo using the YfiN-mediated SCV as a representative strain. The SCV strain exhibited strong, exopolysaccharide-dependent resistance to nematode scavenging and macrophage phagocytosis. Furthermore, the SCV strain effectively persisted over many weeks in mouse infection models, despite exhibiting a marked fitness disadvantage in vitro. Exposure to sub-inhibitory concentrations of antibiotics significantly decreased both the number of suppressors arising, and the relative fitness disadvantage of the SCV mutant in vitro, suggesting that the SCV persistence phenotype may play a more important role during antimicrobial chemotherapy. This study establishes YfiBNR as an important player in P. aeruginosa persistence, and implicates a central role for c-di-GMP, and by extension the SCV phenotype in chronic infections.


Antimicrobial Agents and Chemotherapy | 2014

In Vitro and In Vivo Antibacterial Evaluation of Cadazolid, a New Antibiotic for Treatment of Clostridium difficile Infections

Hans Locher; Peter Seiler; Xinhua Chen; Susanne Schroeder; Philippe Pfaff; Michel Enderlin; Axel Klenk; Elvire Fournier; Christian Hubschwerlen; Daniel Ritz; Ciaran P. Kelly; Wolfgang Keck

ABSTRACT Clostridium difficile is a leading cause of health care-associated diarrhea with significant morbidity and mortality, and new options for the treatment of C. difficile-associated diarrhea (CDAD) are needed. Cadazolid is a new oxazolidinone-type antibiotic that is currently in clinical development for treatment of CDAD. Here, we report the in vitro and in vivo antibacterial evaluation of cadazolid against C. difficile. Cadazolid showed potent in vitro activity against C. difficile with a MIC range of 0.125 to 0.5 μg/ml, including strains resistant to linezolid and fluoroquinolones. In time-kill kinetics experiments, cadazolid showed a bactericidal effect against C. difficile isolates, with >99.9% killing in 24 h, and was more bactericidal than vancomycin. In contrast to metronidazole and vancomycin, cadazolid strongly inhibited de novo toxin A and B formation in stationary-phase cultures of toxigenic C. difficile. Cadazolid also inhibited C. difficile spore formation substantially at growth-inhibitory concentrations. In the hamster and mouse models for CDAD, cadazolid was active, conferring full protection from diarrhea and death with a potency similar to that of vancomycin. These findings support further investigations of cadazolid for the treatment of CDAD.


Antimicrobial Agents and Chemotherapy | 2014

Investigations of the Mode of Action and Resistance Development of Cadazolid, a New Antibiotic for Treatment of Clostridium difficile Infections

Hans Locher; Patrick Caspers; Thierry Bruyère; Susanne Schroeder; Philippe Pfaff; Andreja Knezevic; Wolfgang Keck; Daniel Ritz

ABSTRACT Cadazolid is a new oxazolidinone-type antibiotic currently in clinical development for the treatment of Clostridium difficile-associated diarrhea. Here, we report investigations on the mode of action and the propensity for spontaneous resistance development in C. difficile strains. Macromolecular labeling experiments indicated that cadazolid acts as a potent inhibitor of protein synthesis, while inhibition of DNA synthesis was also observed, albeit only at substantially higher concentrations of the drug. Strong inhibition of protein synthesis was also obtained in strains resistant to linezolid, in agreement with low MICs against such strains. Inhibition of protein synthesis was confirmed in coupled transcription/translation assays using extracts from different C. difficile strains, including strains resistant to linezolid, while inhibitory effects in DNA topoisomerase assays were weak or not detectable under the assay conditions. Spontaneous resistance frequencies of cadazolid were low in all strains tested (generally <10−10 at 2× to 4× the MIC), and in multiple-passage experiments (up to 13 passages) MICs did not significantly increase. Furthermore, no cross-resistance was observed, as cadazolid retained potent activity against strains resistant or nonsusceptible to linezolid, fluoroquinolones, and the new antibiotic fidaxomicin. In conclusion, the data presented here indicate that cadazolid acts primarily by inhibition of protein synthesis, with weak inhibition of DNA synthesis as a potential second mode of action, and suggest a low potential for spontaneous resistance development.


Journal of Medicinal Chemistry | 2013

Design, synthesis, and characterization of novel tetrahydropyran-based bacterial topoisomerase inhibitors with potent anti-gram-positive activity.

Jean-Philippe Surivet; Cornelia Zumbrunn; Georg Rueedi; Christian Hubschwerlen; Daniel Bur; Thierry Bruyère; Hans Locher; Daniel Ritz; Wolfgang Keck; Peter Seiler; Christopher Kohl; Jean-Christophe Gauvin; Azely Mirre; Verena Kaegi; Marina Dos Santos; Mika Gaertner; Jonathan Delers; Michel Enderlin-Paput; Maria Boehme

There is an urgent need for new antibacterial drugs that are effective against infections caused by multidrug-resistant pathogens. Novel nonfluoroquinolone inhibitors of bacterial type II topoisomerases (DNA gyrase and topoisomerase IV) have the potential to become such drugs because they display potent antibacterial activity and exhibit no target-mediated cross-resistance with fluoroquinolones. Bacterial topoisomerase inhibitors that are built on a tetrahydropyran ring linked to a bicyclic aromatic moiety through a syn-diol linker show potent anti-Gram-positive activity, covering isolates with clinically relevant resistance phenotypes. For instance, analog 49c was found to be a dual DNA gyrase-topoisomerase IV inhibitor, with broad antibacterial activity and low propensity for spontaneous resistance development, but suffered from high hERG K(+) channel block. On the other hand, analog 49e displayed lower hERG K(+) channel block while retaining potent in vitro antibacterial activity and acceptable frequency for resistance development. Furthermore, analog 49e showed moderate clearance in rat and promising in vivo efficacy against Staphylococcus aureus in a murine infection model.


Bioorganic & Medicinal Chemistry Letters | 2012

Structure-guided design, synthesis and biological evaluation of novel DNA ligase inhibitors with in vitro and in vivo anti-staphylococcal activity.

Jean-Philippe Surivet; Roland Lange; Christian Hubschwerlen; Wolfgang Keck; Jean-Luc Specklin; Daniel Ritz; Daniel Bur; Hans Locher; Peter Seiler; Daniel S. Strasser; Lars Prade; Christopher Kohl; Christine Schmitt; Gaëlle Chapoux; Eser Ilhan; Nadia Ekambaram; Alcibiade Athanasiou; Andreja Knezevic; Daniela Sabato; Alain Chambovey; Mika Gaertner; Michel Enderlin; Maria Boehme; Virginie Sippel; Pierre Wyss

A series of 2-amino-[1,8]-naphthyridine-3-carboxamides (ANCs) with potent inhibition of bacterial NAD(+)-dependent DNA ligases (LigAs) evolved from a 2,4-diaminopteridine derivative discovered by HTS. The design was guided by several highly resolved X-ray structures of our inhibitors in complex with either Streptococcus pneumoniae or Escherichia coli LigA. The structure-activity-relationship based on the ANC scaffold is discussed. The in-depth characterization of 2-amino-6-bromo-7-(trifluoromethyl)-[1,8]-naphthyridine-3-carboxamide, which displayed promising in vitro (MIC Staphylococcus aureus 1 mg/L) and in vivo anti-staphylococcal activity, is presented.


Journal of Medicinal Chemistry | 2017

Discovery and Optimization of Isoquinoline Ethyl Ureas as Antibacterial Agents

Philippe Panchaud; Thierry Bruyère; Anne-Catherine Blumstein; Daniel Bur; Alain Chambovey; Eric A. Ertel; Markus Gude; Christian Hubschwerlen; Loïc Jacob; Thierry Kimmerlin; Thomas Pfeifer; Lars Prade; Peter Seiler; Daniel Ritz; Georg Rueedi

Our strategy to combat resistant bacteria consisted of targeting the GyrB/ParE ATP-binding sites located on bacterial DNA gyrase and topoisomerase IV and not utilized by marketed antibiotics. Screening around the minimal ethyl urea binding motif led to the identification of isoquinoline ethyl urea 13 as a promising starting point for fragment evolution. The optimization was guided by structure-based design and focused on antibacterial activity in vitro and in vivo, culminating in the discovery of unprecedented substituents able to interact with conserved residues within the ATP-binding site. A detailed characterization of the lead compound highlighted the potential for treatment of the problematic fluoroquinolone-resistant MRSA, VRE, and S. pneumoniae, and the possibility to offer patients an intravenous-to-oral switch therapy was supported by the identification of a suitable prodrug concept. Eventually, hERG K-channel block was identified as the main limitation of this chemical series, and efforts toward its minimization are reported.


Journal of Medicinal Chemistry | 2015

Novel tetrahydropyran-based bacterial topoisomerase inhibitors with potent anti-gram positive activity and improved safety profile.

Jean-Philippe Surivet; Cornelia Zumbrunn; Georg Rueedi; Daniel Bur; Thierry Bruyère; Hans Locher; Daniel Ritz; Peter Seiler; Christopher Kohl; Eric A. Ertel; Patrick Hess; Jean-Christophe Gauvin; Azely Mirre; Verena Kaegi; Marina Dos Santos; Stéphanie Kraemer; Mika Gaertner; Jonathan Delers; Michel Enderlin-Paput; Romain Sube; Hakim Hadana; Wolfgang Keck; Christian Hubschwerlen

Novel antibacterial drugs that are effective against infections caused by multidrug resistant pathogens are urgently needed. In a previous report, we have shown that tetrahydropyran-based inhibitors of bacterial type II topoisomerases (DNA gyrase and topoisomerase IV) display potent antibacterial activity and exhibit no target-mediated cross-resistance with fluoroquinolones. During the course of our optimization program, lead compound 5 was deprioritized due to adverse findings in cardiovascular safety studies. In the effort of mitigating these findings and optimizing further the pharmacological profile of this class of compounds, we have identified a subseries of tetrahydropyran-based molecules that are potent DNA gyrase and topoisomerase IV inhibitors and display excellent antibacterial activity against Gram positive pathogens, including clinically relevant resistant isolates. One representative of this class, compound 32d, elicited only weak inhibition of hERG K(+) channels and hNaV1.5 Na(+) channels, and no effects were observed on cardiovascular parameters in anesthetized guinea pigs. In vivo efficacy in animal infection models has been demonstrated against Staphylococcus aureus and Streptococcus pneumoniae strains.


Chemotherapy | 2010

Dimers of Nostocarboline with Potent Antibacterial Activity

Hans Locher; Daniel Ritz; Philippe Pfaff; Mika Gaertner; Andreja Knezevic; Daniela Sabato; Susanne Schroeder; Damien Barbaras; Karl Gademann

Objectives: In this study, the in vitro antimicrobial activity and spectrum of new dimeric compounds derived from the cyanobacterial alkaloid nostocarboline were investigated. The mechanism of action and selectivity to bacteria were studied and compared to the cationic antiseptic chlorhexidine. Methods:Minimal inhibitory concentrations were determined against clinical isolates and against a panel of microbial reference strains using the CLSI microdilution method. Bacterial membrane damage was addressed by measuring ATP leakage and the mode of action was investigated in Escherichia coli reporter strains. Selectivity was tested by a cytotoxicity assay using MTS. Results: The antimicrobial potency of dimers varied with length of the hydrophobic linker. The most potent compounds, NCD9 and NCD10, had a C10 and C12 linker, respectively, and showed strong activity against Gram-positive bacteria, notably methicillin-resistant Staphylococcus aureus strains. Similar to chlorhexidine, these compounds showed a rapid concentration-dependent bactericidal effect, which correlated with membrane damage as indicated by ATP leakage. NCD9, in contrast to NCD10 and chlorhexidine, lacked activity against yeast strains and showed low cytotoxicity in CHO cells indicating a high degree of selectivity. In E. coli reporter strains, NCD9 induced the DegP response pathway as well as the SOS response, suggesting interaction with both the cell envelope and DNA metabolism. Conclusions: The results presented in this report indicate the potential of this new class of cationic antimicrobial compounds for the design of potent and selective antibacterials with low cytotoxicity.


Antimicrobial Agents and Chemotherapy | 2016

Cadazolid Does Not Promote Intestinal Colonization of Vancomycin-Resistant Enterococci in Mice

Peter Seiler; Michel Enderlin-Paput; Philippe Pfaff; Daniel Ritz; Martine Clozel; Hans Locher

ABSTRACT The promotion of colonization with vancomycin-resistant enterococci (VRE) is one potential side effect during treatment of Clostridium difficile-associated diarrhea (CDAD), resulting from disturbances in gut microbiota. Cadazolid (CDZ) is an investigational antibiotic with potent in vitro activity against C. difficile and against VRE and is currently in clinical development for the treatment of CDAD. We report that CDZ treatment did not lead to intestinal VRE overgrowth in mice.

Collaboration


Dive into the Daniel Ritz's collaboration.

Researchain Logo
Decentralizing Knowledge