Identification of Anti-Mycobacterium and Anti-Legionella Compounds With Potential Distinctive Structural Scaffolds From an HD-PBL Using Phenotypic Screens in Amoebae Host Models

Tubercular Mycobacteria and Legionella pneumophila are the causative agents of potentially fatal respiratory diseases due to their intrinsic pathogenesis but also due to the emergence of antibiotic resistance that limits treatment options. The aim of our study was to explore the antimicrobial activi...

Full description

Bibliographic Details
Main Authors: Nabil Hanna, Sébastien Kicka, Gianpaolo Chiriano, Christopher Harrison, Hajer Ouertatani Sakouhi, Valentin Trofimov, Agata Kranjc, Jahn Nitschke, Marco Pagni, Pierre Cosson, Hubert Hilbi, Leonardo Scapozza, Thierry Soldati
Format: Article
Language:English
Published: Frontiers Media S.A. 2020-02-01
Series:Frontiers in Microbiology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fmicb.2020.00266/full
id doaj-bcff22bba1b245feb81d334c5cb170c5
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Nabil Hanna
Sébastien Kicka
Gianpaolo Chiriano
Christopher Harrison
Hajer Ouertatani Sakouhi
Valentin Trofimov
Agata Kranjc
Jahn Nitschke
Marco Pagni
Pierre Cosson
Hubert Hilbi
Leonardo Scapozza
Thierry Soldati
spellingShingle Nabil Hanna
Sébastien Kicka
Gianpaolo Chiriano
Christopher Harrison
Hajer Ouertatani Sakouhi
Valentin Trofimov
Agata Kranjc
Jahn Nitschke
Marco Pagni
Pierre Cosson
Hubert Hilbi
Leonardo Scapozza
Thierry Soldati
Identification of Anti-Mycobacterium and Anti-Legionella Compounds With Potential Distinctive Structural Scaffolds From an HD-PBL Using Phenotypic Screens in Amoebae Host Models
Frontiers in Microbiology
Mycobacterium
Legionella
anti-infective
ChemGPS
amoebae
author_facet Nabil Hanna
Sébastien Kicka
Gianpaolo Chiriano
Christopher Harrison
Hajer Ouertatani Sakouhi
Valentin Trofimov
Agata Kranjc
Jahn Nitschke
Marco Pagni
Pierre Cosson
Hubert Hilbi
Leonardo Scapozza
Thierry Soldati
author_sort Nabil Hanna
title Identification of Anti-Mycobacterium and Anti-Legionella Compounds With Potential Distinctive Structural Scaffolds From an HD-PBL Using Phenotypic Screens in Amoebae Host Models
title_short Identification of Anti-Mycobacterium and Anti-Legionella Compounds With Potential Distinctive Structural Scaffolds From an HD-PBL Using Phenotypic Screens in Amoebae Host Models
title_full Identification of Anti-Mycobacterium and Anti-Legionella Compounds With Potential Distinctive Structural Scaffolds From an HD-PBL Using Phenotypic Screens in Amoebae Host Models
title_fullStr Identification of Anti-Mycobacterium and Anti-Legionella Compounds With Potential Distinctive Structural Scaffolds From an HD-PBL Using Phenotypic Screens in Amoebae Host Models
title_full_unstemmed Identification of Anti-Mycobacterium and Anti-Legionella Compounds With Potential Distinctive Structural Scaffolds From an HD-PBL Using Phenotypic Screens in Amoebae Host Models
title_sort identification of anti-mycobacterium and anti-legionella compounds with potential distinctive structural scaffolds from an hd-pbl using phenotypic screens in amoebae host models
publisher Frontiers Media S.A.
series Frontiers in Microbiology
issn 1664-302X
publishDate 2020-02-01
description Tubercular Mycobacteria and Legionella pneumophila are the causative agents of potentially fatal respiratory diseases due to their intrinsic pathogenesis but also due to the emergence of antibiotic resistance that limits treatment options. The aim of our study was to explore the antimicrobial activity of a small ligand-based chemical library of 1255 structurally diverse compounds. These compounds were screened in a combination of three assays, two monitoring the intracellular growth of the pathogenic bacteria, Mycobacterium marinum and L. pneumophila, and one assessing virulence of M. marinum. We set up these assays using two amoeba strains, the genetically tractable social amoeba Dictyostelium discoideum and the free-living amoeba Acanthamoeba castellanii. In summary, 64 (5.1%) compounds showed anti-infective/anti-virulence activity in at least one of the three assays. The intracellular assays hit rate varied between 1.7% (n = 22) for M. marinum and 2.8% (n = 35) for L. pneumophila with seven compounds in common for both pathogens. In parallel, 1.2% (n = 15) of the tested compounds were able to restore D. discoideum growth in the presence of M. marinum spiked in a lawn of food bacteria. We also validated the generality of the hits identified in the A. castellanii–M. marinum anti-infective screen using the D. discoideum–M. marinum host–pathogen model. The characterization of anti-infective and antibacterial hits in the latter infection model revealed compounds able to reduce intracellular growth more than 50% at 30 μM. Moreover, the chemical space and physico-chemical properties of the anti-M. marinum hits were compared to standard and candidate Mycobacterium tuberculosis (Mtb) drugs using ChemGPS-NP. A principle component analysis identified separate clusters for anti-M. marinum and anti-L. pneumophila hits unveiling the potentially new physico-chemical properties of these hits compared to standard and candidate M. tuberculosis drugs. Our studies underscore the relevance of using a combination of low-cost and low-complexity assays with full 3R compliance in concert with a rationalized focused library of compounds to identify new chemical scaffolds and to dissect some of their properties prior to taking further steps toward compound development.
topic Mycobacterium
Legionella
anti-infective
ChemGPS
amoebae
url https://www.frontiersin.org/article/10.3389/fmicb.2020.00266/full
work_keys_str_mv AT nabilhanna identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
AT sebastienkicka identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
AT gianpaolochiriano identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
AT christopherharrison identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
AT hajerouertatanisakouhi identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
AT valentintrofimov identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
AT agatakranjc identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
AT jahnnitschke identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
AT marcopagni identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
AT pierrecosson identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
AT huberthilbi identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
AT leonardoscapozza identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
AT thierrysoldati identificationofantimycobacteriumandantilegionellacompoundswithpotentialdistinctivestructuralscaffoldsfromanhdpblusingphenotypicscreensinamoebaehostmodels
_version_ 1724913892323229696
spelling doaj-bcff22bba1b245feb81d334c5cb170c52020-11-25T02:11:36ZengFrontiers Media S.A.Frontiers in Microbiology1664-302X2020-02-011110.3389/fmicb.2020.00266510767Identification of Anti-Mycobacterium and Anti-Legionella Compounds With Potential Distinctive Structural Scaffolds From an HD-PBL Using Phenotypic Screens in Amoebae Host ModelsNabil Hanna0Sébastien Kicka1Gianpaolo Chiriano2Christopher Harrison3Hajer Ouertatani Sakouhi4Valentin Trofimov5Agata Kranjc6Jahn Nitschke7Marco Pagni8Pierre Cosson9Hubert Hilbi10Leonardo Scapozza11Thierry Soldati12Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, SwitzerlandDepartment of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, SwitzerlandPharmaceutical Biochemistry/Chemistry, School of Pharmaceutical Sciences, University of Geneva, Geneva, SwitzerlandMax von Pettenkofer Institute, Ludwig Maximilian University of Munich, Munich, GermanyDepartment of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, SwitzerlandDepartment of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, SwitzerlandPharmaceutical Biochemistry/Chemistry, School of Pharmaceutical Sciences, University of Geneva, Geneva, SwitzerlandDepartment of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, SwitzerlandSwiss Institute of Bioinformatics, Lausanne, SwitzerlandDepartment of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, SwitzerlandInstitute of Medical Microbiology, University of Zurich, Zurich, SwitzerlandPharmaceutical Biochemistry/Chemistry, School of Pharmaceutical Sciences, University of Geneva, Geneva, SwitzerlandDepartment of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, SwitzerlandTubercular Mycobacteria and Legionella pneumophila are the causative agents of potentially fatal respiratory diseases due to their intrinsic pathogenesis but also due to the emergence of antibiotic resistance that limits treatment options. The aim of our study was to explore the antimicrobial activity of a small ligand-based chemical library of 1255 structurally diverse compounds. These compounds were screened in a combination of three assays, two monitoring the intracellular growth of the pathogenic bacteria, Mycobacterium marinum and L. pneumophila, and one assessing virulence of M. marinum. We set up these assays using two amoeba strains, the genetically tractable social amoeba Dictyostelium discoideum and the free-living amoeba Acanthamoeba castellanii. In summary, 64 (5.1%) compounds showed anti-infective/anti-virulence activity in at least one of the three assays. The intracellular assays hit rate varied between 1.7% (n = 22) for M. marinum and 2.8% (n = 35) for L. pneumophila with seven compounds in common for both pathogens. In parallel, 1.2% (n = 15) of the tested compounds were able to restore D. discoideum growth in the presence of M. marinum spiked in a lawn of food bacteria. We also validated the generality of the hits identified in the A. castellanii–M. marinum anti-infective screen using the D. discoideum–M. marinum host–pathogen model. The characterization of anti-infective and antibacterial hits in the latter infection model revealed compounds able to reduce intracellular growth more than 50% at 30 μM. Moreover, the chemical space and physico-chemical properties of the anti-M. marinum hits were compared to standard and candidate Mycobacterium tuberculosis (Mtb) drugs using ChemGPS-NP. A principle component analysis identified separate clusters for anti-M. marinum and anti-L. pneumophila hits unveiling the potentially new physico-chemical properties of these hits compared to standard and candidate M. tuberculosis drugs. Our studies underscore the relevance of using a combination of low-cost and low-complexity assays with full 3R compliance in concert with a rationalized focused library of compounds to identify new chemical scaffolds and to dissect some of their properties prior to taking further steps toward compound development.https://www.frontiersin.org/article/10.3389/fmicb.2020.00266/fullMycobacteriumLegionellaanti-infectiveChemGPSamoebae