1
|
Braun E, Andany SH, Kangül M, Asmari NS, McKinney JD, Fantner GE. A hermetically closed sample chamber enables time-lapse nano-characterization of pathogenic microorganisms in vitro. NANOSCALE ADVANCES 2025; 7:2290-2300. [PMID: 40041386 PMCID: PMC11873737 DOI: 10.1039/d4na01053a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/13/2025] [Indexed: 03/06/2025]
Abstract
Pathogenic microorganisms, such as pathogenic mycobacteria, pose a global health burden. Studying these organisms is crucial for gaining detailed knowledge about the pathogens and the diseases they cause. To handle pathogenic organisms, specific biosafety measures appropriate to the virulence of the organism must be fulfilled, most importantly ensuring that all manipulations of pathogenic material are performed within a confined environment. Atomic force microscopy (AFM) is a powerful technique to study biological samples at nanometer-scale resolution, yielding also mechanical properties, all while maintaining physiological conditions. However, standard AFM sample holders do not meet stringent biosafety requirements since they do not constitute a confined system. AFM imaging relies on direct contact between the cantilever and the sample and is sensitive to mechanical interference, rendering conventional containment systems for handling infectious substances inapplicable. Here, we introduce a hermetically sealed AFM sample chamber that meets biosafety demands while satisfying the mechanical and optical constraints of correlated optical microscopy and AFM. We imaged various pathogenic mycobacteria to demonstrate the chamber's versatility and effectiveness in containing biohazardous materials. This sample chamber enables high-resolution, time-lapse correlated imaging and biomechanical characterization of pathogenic microorganisms in vitro. It broadens the scope of research with pathogenic microorganisms under safe and controlled conditions.
Collapse
Affiliation(s)
- Esther Braun
- School of Engineering, Swiss Federal Institute of Technology (EPFL) Lausanne Switzerland
- School of Life Sciences, Swiss Federal Institute of Technology (EPFL) Lausanne Switzerland
| | - Santiago H Andany
- School of Engineering, Swiss Federal Institute of Technology (EPFL) Lausanne Switzerland
| | - Mustafa Kangül
- School of Engineering, Swiss Federal Institute of Technology (EPFL) Lausanne Switzerland
| | - Navid S Asmari
- School of Engineering, Swiss Federal Institute of Technology (EPFL) Lausanne Switzerland
| | - John D McKinney
- School of Engineering, Swiss Federal Institute of Technology (EPFL) Lausanne Switzerland
| | - Georg E Fantner
- School of Engineering, Swiss Federal Institute of Technology (EPFL) Lausanne Switzerland
| |
Collapse
|
2
|
Nitschke J, Huber R, Vossio S, Moreau D, Marcourt L, Gindro K, Queiroz EF, Soldati T, Hanna N. Discovery of anti-infective compounds against Mycobacterium marinum after biotransformation of simple natural stilbenes by a fungal secretome. Front Microbiol 2024; 15:1439814. [PMID: 39355425 PMCID: PMC11443511 DOI: 10.3389/fmicb.2024.1439814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/29/2024] [Indexed: 10/03/2024] Open
Abstract
Introduction Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, remains a serious threat to human health worldwide and the quest for new anti-tubercular drugs is an enduring and demanding journey. Natural products (NPs) have played a significant role in advancing drug therapy of infectious diseases. Methods This study evaluated the suitability of a high-throughput infection system composed of the host amoeba Dictyostelium discoideum (Dd) and Mycobacterium marinum (Mm), a close relative of Mtb, to identify anti-infective compounds. Growth of Dd and intracellular Mm were quantified by using luminescence and fluorescence readouts in phenotypic assays. The system was first benchmarked with a set of therapeutic anti-Mtb antibiotics and then used to screen a library of biotransformed stilbenes. Results The study confirmed both efficacy of established antibiotics such as rifampicin and bedaquiline, with activities below defined anti-mycobacterium susceptibility breakpoints, and the lack of activity of pyrazinamide against Mm. The screening revealed the promising anti-infective activities of trans-δ-viniferins and in particular of two compounds 17 and 19 with an IC50 of 18.1 μM, 9 μM, respectively. Both compounds had no activity on Mm in broth. Subsequent exploration via halogenation and structure-activity relationship studies led to the identification of derivatives with improved selectivity and potency. The modes of action of the anti-infective compounds may involve inhibition of mycobacterial virulence factors or boosting of host defense. Discussion The study highlights the potential of biotransformation and NP-inspired derivatization approaches for drug discovery and underscores the utility of the Dd-Mm infection system in identifying novel anti-infective compounds.
Collapse
Affiliation(s)
- Jahn Nitschke
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Robin Huber
- School of Pharmaceutical Sciences, University of Geneva, CMU, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, Geneva, Switzerland
| | - Stefania Vossio
- ACCESS Screening Platform, NCCR Chemical Biology, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Dimitri Moreau
- ACCESS Screening Platform, NCCR Chemical Biology, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Laurence Marcourt
- School of Pharmaceutical Sciences, University of Geneva, CMU, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, Geneva, Switzerland
| | - Katia Gindro
- Mycology Group, Research Department Plant Protection, Agroscope, Nyon, Switzerland
| | - Emerson F. Queiroz
- School of Pharmaceutical Sciences, University of Geneva, CMU, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, Geneva, Switzerland
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Nabil Hanna
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| |
Collapse
|
3
|
Anand A, Mazur AC, Rosell-Arevalo P, Franzkoch R, Breitsprecher L, Listian SA, Hüttel SV, Müller D, Schäfer DG, Vormittag S, Hilbi H, Maniak M, Gutierrez MG, Barisch C. ER-dependent membrane repair of mycobacteria-induced vacuole damage. mBio 2023; 14:e0094323. [PMID: 37676004 PMCID: PMC10653851 DOI: 10.1128/mbio.00943-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/13/2023] [Indexed: 09/08/2023] Open
Abstract
IMPORTANCE Tuberculosis still remains a global burden and is one of the top infectious diseases from a single pathogen. Mycobacterium tuberculosis, the causative agent, has perfected many ways to replicate and persist within its host. While mycobacteria induce vacuole damage to evade the toxic environment and eventually escape into the cytosol, the host recruits repair machineries to restore the MCV membrane. However, how lipids are delivered for membrane repair is poorly understood. Using advanced fluorescence imaging and volumetric correlative approaches, we demonstrate that this involves the recruitment of the endoplasmic reticulum (ER)-Golgi lipid transfer protein OSBP8 in the Dictyostelium discoideum/Mycobacterium marinum system. Strikingly, depletion of OSBP8 affects lysosomal function accelerating mycobacterial growth. This indicates that an ER-dependent repair pathway constitutes a host defense mechanism against intracellular pathogens such as M. tuberculosis.
Collapse
Affiliation(s)
- Aby Anand
- Division of Molecular Infection Biology, Department of Biology & Center of Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany
| | - Anna-Carina Mazur
- Division of Molecular Infection Biology, Department of Biology & Center of Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany
| | - Patricia Rosell-Arevalo
- Host–Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Rico Franzkoch
- Integrated Bioimaging Facility, Center of Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany
| | - Leonhard Breitsprecher
- Integrated Bioimaging Facility, Center of Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany
| | - Stevanus A. Listian
- Division of Molecular Infection Biology, Department of Biology & Center of Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany
| | - Sylvana V. Hüttel
- Division of Molecular Infection Biology, Department of Biology & Center of Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany
| | - Danica Müller
- Division of Molecular Infection Biology, Department of Biology & Center of Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany
| | - Deise G. Schäfer
- Division of Molecular Infection Biology, Department of Biology & Center of Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany
| | - Simone Vormittag
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| | - Hubert Hilbi
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| | - Markus Maniak
- Department of Cell Biology, University of Kassel, Kassel, Germany
| | - Maximiliano G. Gutierrez
- Host–Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Caroline Barisch
- Division of Molecular Infection Biology, Department of Biology & Center of Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany
- Centre for Structural Systems Biology, Hamburg, Germany
- Division of Host-Microbe Interactome, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| |
Collapse
|
4
|
Cha GY, Seo H, Oh J, Kim BJ, Kim BJ. Potential Use of Mycobacterium paragordonae for Antimycobacterial Drug Screening Systems. J Microbiol 2023; 61:121-129. [PMID: 36719620 DOI: 10.1007/s12275-022-00009-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/20/2022] [Accepted: 11/25/2022] [Indexed: 02/01/2023]
Abstract
Our recent genome-based study indicated that Mycobacterium paragordonae (Mpg) has evolved to become more adapted to an intracellular lifestyle within free-living environmental amoeba and its enhanced intracellular survival within Acanthamoeba castellanii was also proved. Here, we sought to investigate potential use of Mpg for antimycobacterial drug screening systems. Our data showed that Mpg is more susceptible to various antibiotics compared to the close species M. marinum (Mmar) and M. gordonae, further supporting its intracellular lifestyle in environments, which would explain its protection from environmental insults. In addition, we developed two bacterial whole-cell-based drug screening systems using a recombinant Mpg stain harboring a luciferase reporter vector (rMpg-LuxG13): one for direct application to rMpg-LuxG13 and the other for drug screening via the interaction of rMpg-LuxG13 with A. castellanii. Direct application to rMpg-LuxG13 showed lower inhibitory concentration 50 (IC50) values of rifampin, isoniazid, clarithromycin, and ciprofloxacin against Mpg compared to Mmar. Application of drug screening system via the interaction of rMpg-LuxG13 with A. castellanii also exhibited lower IC50 values for rifampin against Mpg compared to Mmar. In conclusion, our data indicate that Mpg is more susceptible to various antibiotics than other strains. In addition, our data also demonstrate the feasibility of two whole cell-based drug screening systems using rMpg-LuxG13 strain for the discovery of novel anti-mycobacterial drugs.
Collapse
Affiliation(s)
- Ga-Yeong Cha
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- BK21 Four Biomedical Science Project, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hyejun Seo
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jaehun Oh
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- BK21 Four Biomedical Science Project, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Byoung-Jun Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, Republic of Korea.
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea.
- Liver Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea.
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea.
- Institute of Endemic Diseases, Seoul National University Medical Research Center (SNUMRC), Seoul, Republic of Korea.
- BK21 Four Biomedical Science Project, College of Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Protozoal food vacuoles enhance transformation in Vibrio cholerae through SOS-regulated DNA integration. THE ISME JOURNAL 2022; 16:1993-2001. [PMID: 35577916 PMCID: PMC9296650 DOI: 10.1038/s41396-022-01249-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 04/20/2022] [Accepted: 04/29/2022] [Indexed: 11/08/2022]
Abstract
Vibrio cholerae, the bacterial pathogen responsible for the diarrheal disease cholera, resides in the aquatic environment between outbreaks. For bacteria, genetic variation by lateral gene transfer (LGT) is important for survival and adaptation. In the aquatic environment, V. cholerae is predominantly found in biofilms associated with chitinous organisms or with chitin "rain". Chitin induces competency in V. cholerae, which can lead to LGT. In the environment, V. cholerae is also subjected to predation pressure by protist. Here we investigated whether protozoal predation affected LGT using the integron as a model. Integrons facilitate the integration of mobile DNA (gene cassettes) into the bacterial chromosome. We report that protozoal predation enhances transformation of a gene cassette by as much as 405-fold. We show that oxidative radicals produced in the protozoal phagosome induces the universal SOS response, which in turn upregulates the integron-integrase, the recombinase that facilitates cassette integration. Additionally, we show that during predation, V. cholerae requires the type VI secretion system to acquire the gene cassette from Escherichia coli. These results show that protozoal predation enhances LGT thus producing genetic variants that may have increased capacity to survive grazing. Additionally, the conditions in the food vacuole may make it a "hot spot" for LGT by accumulating diverse bacteria and inducing the SOS response helping drive genetic diversification and evolution.
Collapse
|
6
|
Amaro F, Martín-González A. Microbial warfare in the wild-the impact of protists on the evolution and virulence of bacterial pathogens. Int Microbiol 2021; 24:559-571. [PMID: 34365574 DOI: 10.1007/s10123-021-00192-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/03/2021] [Accepted: 06/28/2021] [Indexed: 01/01/2023]
Abstract
During the long history of co-evolution with protists, bacteria have evolved defense strategies to avoid grazing and survive phagocytosis. These mechanisms allow bacteria to exploit phagocytic cells as a protective niche in which to escape from environmental stress and even replicate. Importantly, these anti-grazing mechanisms can function as virulence factors when bacteria infect humans. Here, we discuss how protozoan predation exerts a selective pressure driving bacterial virulence and shaping their genomes, and how bacteria-protist interactions might contribute to the spread of antibiotic resistance as well. We provide examples to demonstrate that besides being voracious bacterial predators, protozoa can serve as melting pots where intracellular organisms exchange genetic information, or even "training grounds" where some pathogens become hypervirulent after passing through. In this special issue, we would like to emphasize the tremendous impact of bacteria-protist interactions on human health and the potential of amoebae as model systems to study biology and evolution of a variety of pathogens. Besides, a better understanding of bacteria-protist relationships will help us expand our current understanding of bacterial virulence and, likely, how pathogens emerge.
Collapse
Affiliation(s)
- Francisco Amaro
- Department of Genetics, Physiology and Microbiology, School of Biology, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Ana Martín-González
- Department of Genetics, Physiology and Microbiology, School of Biology, Complutense University of Madrid, 28040, Madrid, Spain
| |
Collapse
|
7
|
Subhash N, Sundaramurthy V. Advances in host-based screening for compounds with intracellular anti-mycobacterial activity. Cell Microbiol 2021; 23:e13337. [PMID: 33813790 DOI: 10.1111/cmi.13337] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/14/2022]
Abstract
Intracellular pathogens interact with host systems in intimate ways to sustain a pathogenic lifestyle. Consequently, these interactions can potentially be targets of host-directed interventions against infectious diseases. In case of tuberculosis (TB), caused by the bacterium Mycobacterium tuberculosis (Mtb), while effective anti-tubercular compounds are available, the long treatment duration and emerging drug resistance necessitate identification of new class of molecules with anti-TB activity, as well as new treatment strategies. A significant part of the effort in finding new anti-TB drugs is focused on bacterial targets in bacterial systems. However, the host environment plays a major role in pathogenesis mechanisms and must be considered actively in these efforts. On the one hand, the bacterial origin targets must be relevant and accessible in the host, while on the other hand, new host origin targets required for the bacterial survival can be targeted. Such targets are good candidates for host-directed therapeutics, a strategy gaining traction as an adjunct in TB treatment. In this review, we will summarise the screening platforms used to identify compounds with anti-tubercular activities inside different host environments and outline recent technical advances in these platforms. Finally, while the examples given are specific to mycobacteria, the methods and principles outlined are broadly applicable to most intracellular infections.
Collapse
Affiliation(s)
- Neeraja Subhash
- National Center for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India.,SASTRA University, Thanjavur, India
| | | |
Collapse
|
8
|
Guillonneau R, Baraquet C, Molmeret M. Marine Bacteria Display Different Escape Mechanisms When Facing Their Protozoan Predators. Microorganisms 2020; 8:microorganisms8121982. [PMID: 33322808 PMCID: PMC7763514 DOI: 10.3390/microorganisms8121982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/24/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022] Open
Abstract
Free-living amoeba are members of microbial communities such as biofilms in terrestrial, fresh, and marine habitats. Although they are known to live in close association with bacteria in many ecosystems such as biofilms, they are considered to be major bacterial predators in many ecosystems. Little is known on the relationship between protozoa and marine bacteria in microbial communities, more precisely on how bacteria are able survive in environmental niches where these bacterial grazers also live. The objective of this work is to study the interaction between the axenized ubiquitous amoeba Acanthamoeba castellanii and four marine bacteria isolated from immersed biofilm, in order to evaluate if they would be all grazed upon by amoeba or if they would be able to survive in the presence of their predator. At a low bacteria-to-amoeba ratio, we show that each bacterium is phagocytized and follows a singular intracellular path within this host cell, which appears to delay or to prevent bacterial digestion. In particular, one of the bacteria was found in the amoeba nucleolar compartment whereas another strain was expelled from the amoeba in vesicles. We then looked at the fate of the bacteria grown in a higher bacteria-to-amoeba ratio, as a preformed mono- or multi-species biofilm in the presence of A. castellanii. We show that all biofilms were subjected to detachment from the surface in the presence of the amoeba or its supernatant. Overall, these results show that bacteria, when facing the same predator, exhibit a variety of escape mechanisms at the cellular and population level, when we could have expected a simple bacterial grazing. Therefore, this study unravels new insights into the survival of environmental bacteria when facing predators that they could encounter in the same microbial communities.
Collapse
Affiliation(s)
- Richard Guillonneau
- Laboratoire MAPIEM, EA4323, Université de Toulon, 83130 La Garde, France; (R.G.); (C.B.)
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Claudine Baraquet
- Laboratoire MAPIEM, EA4323, Université de Toulon, 83130 La Garde, France; (R.G.); (C.B.)
| | - Maëlle Molmeret
- Laboratoire MAPIEM, EA4323, Université de Toulon, 83130 La Garde, France; (R.G.); (C.B.)
- Correspondence:
| |
Collapse
|
9
|
Rampacci E, Stefanetti V, Passamonti F, Henao-Tamayo M. Preclinical Models of Nontuberculous Mycobacteria Infection for Early Drug Discovery and Vaccine Research. Pathogens 2020; 9:E641. [PMID: 32781698 PMCID: PMC7459799 DOI: 10.3390/pathogens9080641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022] Open
Abstract
Nontuberculous mycobacteria (NTM) represent an increasingly prevalent etiology of soft tissue infections in animals and humans. NTM are widely distributed in the environment and while, for the most part, they behave as saprophytic organisms, in certain situations, they can be pathogenic, so much so that the incidence of NTM infections has surpassed that of Mycobacterium tuberculosis in developed countries. As a result, a growing body of the literature has focused attention on the critical role that drug susceptibility tests and infection models play in the design of appropriate therapeutic strategies against NTM diseases. This paper is an overview of the in vitro and in vivo models of NTM infection employed in the preclinical phase for early drug discovery and vaccine development. It summarizes alternative methods, not fully explored, for the characterization of anti-mycobacterial compounds.
Collapse
Affiliation(s)
- Elisa Rampacci
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (E.R.); (V.S.)
| | - Valentina Stefanetti
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (E.R.); (V.S.)
| | - Fabrizio Passamonti
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (E.R.); (V.S.)
| | - Marcela Henao-Tamayo
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA;
| |
Collapse
|
10
|
Hanna N, Kicka S, Chiriano G, Harrison C, Sakouhi HO, Trofimov V, Kranjc A, Nitschke J, Pagni M, Cosson P, Hilbi H, Scapozza L, Soldati T. Identification of Anti- Mycobacterium and Anti- Legionella Compounds With Potential Distinctive Structural Scaffolds From an HD-PBL Using Phenotypic Screens in Amoebae Host Models. Front Microbiol 2020; 11:266. [PMID: 32153546 PMCID: PMC7047896 DOI: 10.3389/fmicb.2020.00266] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/05/2020] [Indexed: 12/22/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Nabil Hanna
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Sébastien Kicka
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Gianpaolo Chiriano
- Pharmaceutical Biochemistry/Chemistry, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Christopher Harrison
- Max von Pettenkofer Institute, Ludwig Maximilian University of Munich, Munich, Germany
| | - Hajer Ouertatani Sakouhi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Valentin Trofimov
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Agata Kranjc
- Pharmaceutical Biochemistry/Chemistry, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Jahn Nitschke
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Marco Pagni
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Pierre Cosson
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Hubert Hilbi
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry/Chemistry, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| |
Collapse
|
11
|
Knobloch P, Koliwer-Brandl H, Arnold FM, Hanna N, Gonda I, Adenau S, Personnic N, Barisch C, Seeger MA, Soldati T, Hilbi H. Mycobacterium marinum produces distinct mycobactin and carboxymycobactin siderophores to promote growth in broth and phagocytes. Cell Microbiol 2020; 22:e13163. [PMID: 31945239 DOI: 10.1111/cmi.13163] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/22/2019] [Indexed: 02/06/2023]
Abstract
Mycobacterium marinum is a model organism for pathogenic Mycobacterium species, including Mycobacterium tuberculosis, the causative agent of tuberculosis. These pathogens enter phagocytes and replicate within the Mycobacterium-containing vacuole, possibly followed by vacuole exit and growth in the host cell cytosol. Mycobacteria release siderophores called mycobactins to scavenge iron, an essential yet poorly soluble and available micronutrient. To investigate the role of M. marinum mycobactins, we purified by organic solvent extraction and identified by mass spectrometry the lipid-bound mycobactin (MBT) and the water-soluble variant carboxymycobactin (cMBT). Moreover, we generated by specialised phage transduction a defined M. marinum ΔmbtB deletion mutant predicted to be defective for mycobactin production. The M. marinum ΔmbtB mutant strain showed a severe growth defect in broth and phagocytes, which was partially complemented by supplying the mbtB gene on a plasmid. Furthermore, purified Fe-MBT or Fe-cMBT improved the growth of wild type as well as ΔmbtB mutant bacteria on minimal plates, but only Fe-cMBT promoted the growth of wild-type M. marinum during phagocyte infection. Finally, the intracellular growth of M. marinum ΔmbtB in Acanthamoeba castellanii amoebae was restored by coinfection with wild-type bacteria. Our study identifies and characterises the M. marinum MBT and cMBT siderophores and reveals the requirement of mycobactins for extra- and intracellular growth of the pathogen.
Collapse
Affiliation(s)
- Paulina Knobloch
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| | | | - Fabian M Arnold
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| | - Nabil Hanna
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Imre Gonda
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| | - Sophia Adenau
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| | - Nicolas Personnic
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| | - Caroline Barisch
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Markus A Seeger
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Hubert Hilbi
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
12
|
Sharma M, Tyagi JL, Poluri KM. Quantifying bacterial cell lysis using GFP based fluorimetric assay. Int J Biol Macromol 2019; 138:881-889. [PMID: 31356938 DOI: 10.1016/j.ijbiomac.2019.07.172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 11/24/2022]
Abstract
Quantitative measurement of cell lysis against a given microbial strain is essential to calculate the antimicrobial potency of protein/peptide/nanomaterial based formulations. Fluorescence spectroscopy based measurements offer precise quantification of a process via selected flurophore emission profile. In this context, we elucidate a reliable and robust green fluorescent protein (GFP) based fluorescence spectroscopy protocol to evaluate the antimicrobial activity of proteins. The technique is based on the fact that the intensity of the GFP emission released from cells correlates with cell lysis and henceforth the antimicrobial potential of the chosen agent. The technique was demonstrated with two different families of bacteriophage endolysins (T7 and T4 endolysins) using GFP expressing E. coli cells. The GFP based method allowed the absolute quantification of T4 and T7 endolysins cell lysis characteristics at different pH, salt concentrations, and metal ions. The results obtained from GFP based fluorimetric assay were substantiated with turbidimetric assay and fluorescence microscopy. This fluorimetric method in conjugation with different GFP expressing microbial strains and antimicrobial agents can be efficiently applied as a quantification technique to precisely measure cell lysis.
Collapse
Affiliation(s)
- Meenakshi Sharma
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Jaya Lakshmi Tyagi
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Krishna Mohan Poluri
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| |
Collapse
|
13
|
Diop EA, Jacquat J, Drouin N, Queiroz EF, Wolfender JL, Diop T, Schappler J, Rudaz S. Quantitative CE analysis of punicalagin in Combretum aculeatum extracts traditionally used in Senegal for the treatment of tuberculosis. Electrophoresis 2019; 40:2820-2827. [PMID: 31407800 DOI: 10.1002/elps.201900240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 07/26/2019] [Accepted: 08/10/2019] [Indexed: 12/12/2022]
Abstract
Mycobacterium tuberculosis is the causative agent of tuberculosis, an infectious bacterial disease, which most commonly affects the lungs. In the search for novel active compounds or medicines against tuberculosis, an ethnopharmacological survey combined with a host-pathogen assay has recently highlighted the potency of an aqueous extract of Combretum aculeatum. C. aculeatum is used in traditional medicine and has demonstrated a significant in vitro antimycobacterial activity. Punicalagin, an ellagitannin, was isolated and found to be related to the biological activity of the extract. An analytical method for the evaluation of punicalagin in C. aculeatum was developed by capillary electrophoresis. After method optimization, the quantification of punicalagin was achieved for the evaluation of various plant extracts to determine the content of punicalagin related to the extraction modes and conditions, origin of the plant material, and harvesting period. The developed method demonstrated that the leaves presented the highest punicalagin content compared to the seeds and stems. A decoction of 30 min in boiling water was found to be the best extraction mode of C. aculeatum.
Collapse
Affiliation(s)
- ElHadji Assane Diop
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland.,Biology Department, Université Cheikh Anta Diop, Dakar, Senegal
| | - Jenna Jacquat
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Nicolas Drouin
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Emerson Ferreira Queiroz
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Jean-Luc Wolfender
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Tahir Diop
- Biology Department, Université Cheikh Anta Diop, Dakar, Senegal
| | - Julie Schappler
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Serge Rudaz
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| |
Collapse
|
14
|
Diop EHA, Queiroz EF, Marcourt L, Kicka S, Rudaz S, Diop T, Soldati T, Wolfender JL. Antimycobacterial activity in a single-cell infection assay of ellagitannins from Combretum aculeatum and their bioavailable metabolites. JOURNAL OF ETHNOPHARMACOLOGY 2019; 238:111832. [PMID: 30914349 DOI: 10.1016/j.jep.2019.111832] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 03/08/2019] [Accepted: 03/20/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The water decoction of Combretum aculeatum aerial parts is traditionally used in Senegal to treat tuberculosis (TB). The extract shows significant antimycobacterial activity in a validated single-cell infection assay. AIM OF THE STUDY The main aim of this study was to identify the antimycobacterial compounds in the water decoction of Combretum aculeatum. Since the traditional preparations are used orally, a bioactivity assessment of the possible bioavailable human metabolites was also performed. MATERIALS AND METHODS The Combretum aculeatum water decoction extract was first fractionated by flash chromatography. The fractions were submitted to an antibiotic assay against Mycobacterium marinum and to a single-cell infection assay involving Acanthamoeba castellanii as a host. Using these approaches, it was possible to correlate the antimycobacterial activity with two zones of the chromatogram. In parallel with this liquid chromatography (LC)-based activity profiling, high-resolution mass spectrometry (UHPLC-HRMS/MS) revealed the presence of ellagitannin (Et) derivatives in the active zones of the chromatogram. Isolation of the active compounds was performed by preparative chromatography. The structures of the isolated compounds were elucidated by nuclear magnetic resonance (NMR). Additionally, the main human metabolites of commercially available Ets were biologically evaluated in a similar manner. RESULTS The in vitro bioassay-guided isolation of the Combretum aculeatum water extract led to the identification of three Ets (1-3) and ellagic acid (4). The major compounds 2 and 3 (α- and β-punicalagin, respectively), exhibited anti-infective activity with an IC50 of 51.48 μM. In view of the documented intestinal metabolism of these compounds, some metabolites, namely, urolithin A (5), urolithin B (6) and urolithin D (7), were investigated for their antimycobacterial activity in the two assays. Urolithin D (7) exhibited the strongest anti-infective activity, with an IC50 of 345.50 μM, but this was moderate compared to the positive control rifampin (IC50 of 6.99 μM). The compounds assayed had no observable cytotoxicity towards the amoeba host cells at concentrations lower than 200 μg/mL. CONCLUSION The observed antimycobacterial properties of the traditional water decoction of Combretum aculeatum might be related to the activity of Ets derivatives (1-3) and their metabolites, such as ellagic acid (4) and urolithin D (7). Despite the relatively weak activity of these metabolites, the high consumption of tannins achieved by taking the usual traditional decoction doses should lead to an important increase in the plasmatic concentrations of these active and bioavailable metabolites. These results support to some extent the traditional use of Combretum aculeatum to treat tuberculosis.
Collapse
Affiliation(s)
- El Hadji Assane Diop
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211, Geneva 11, Switzerland; Biology Department, University Cheikh Anta Diop, Dakar, Senegal
| | - Emerson Ferreira Queiroz
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211, Geneva 11, Switzerland
| | - Laurence Marcourt
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211, Geneva 11, Switzerland
| | - Sébastien Kicka
- Department of Biochemistry, Faculty of Science, University of Geneva, Quai Ansermet 30, 1211, Geneva 4, Switzerland
| | - Serge Rudaz
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211, Geneva 11, Switzerland
| | - Tahir Diop
- Biology Department, University Cheikh Anta Diop, Dakar, Senegal
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Science, University of Geneva, Quai Ansermet 30, 1211, Geneva 4, Switzerland
| | - Jean-Luc Wolfender
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211, Geneva 11, Switzerland.
| |
Collapse
|
15
|
Thewes S, Soldati T, Eichinger L. Editorial: Amoebae as Host Models to Study the Interaction With Pathogens. Front Cell Infect Microbiol 2019; 9:47. [PMID: 30941316 PMCID: PMC6433779 DOI: 10.3389/fcimb.2019.00047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 02/13/2019] [Indexed: 01/01/2023] Open
Affiliation(s)
- Sascha Thewes
- Department of Biology, Chemistry, Pharmacy, Institute for Biology - Microbiology, Freie Universität Berlin, Berlin, Germany
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Science, University of Geneva, Sciences II, Geneva, Switzerland
| | - Ludwig Eichinger
- Medical Faculty, Center for Biochemistry, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
16
|
Koliwer‐Brandl H, Knobloch P, Barisch C, Welin A, Hanna N, Soldati T, Hilbi H. DistinctMycobacterium marinumphosphatases determine pathogen vacuole phosphoinositide pattern, phagosome maturation, and escape to the cytosol. Cell Microbiol 2019; 21:e13008. [DOI: 10.1111/cmi.13008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 12/18/2018] [Accepted: 01/12/2019] [Indexed: 01/02/2023]
Affiliation(s)
- Hendrik Koliwer‐Brandl
- Faculty of Medicine, Institute of Medical MicrobiologyUniversity of Zurich Zurich Switzerland
| | - Paulina Knobloch
- Faculty of Medicine, Institute of Medical MicrobiologyUniversity of Zurich Zurich Switzerland
| | - Caroline Barisch
- Faculty of Science, Department of BiochemistryUniversity of Geneva Geneva Switzerland
| | - Amanda Welin
- Faculty of Medicine, Institute of Medical MicrobiologyUniversity of Zurich Zurich Switzerland
| | - Nabil Hanna
- Faculty of Science, Department of BiochemistryUniversity of Geneva Geneva Switzerland
| | - Thierry Soldati
- Faculty of Science, Department of BiochemistryUniversity of Geneva Geneva Switzerland
| | - Hubert Hilbi
- Faculty of Medicine, Institute of Medical MicrobiologyUniversity of Zurich Zurich Switzerland
| |
Collapse
|
17
|
Dubois V, Laencina L, Bories A, Le Moigne V, Pawlik A, Herrmann JL, Girard-Misguich F. Identification of Virulence Markers of Mycobacterium abscessus for Intracellular Replication in Phagocytes. J Vis Exp 2018. [PMID: 30320743 DOI: 10.3791/57766] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
What differentiates Mycobacterium abscessus from other saprophytic mycobacteria is the ability to resist phagocytosis by human macrophages and the ability to multiply inside such cells. These virulence traits render M. abscessus pathogenic, especially in vulnerable hosts with underlying structural lung disease, such as cystic fibrosis, bronchiectasis or tuberculosis. How patients become infected with M. abscessus remains unclear. Unlike many mycobacteria, M. abscessus is not found in the environment but might reside inside amoebae, environmental phagocytes that represent a potential reservoir for M. abscessus. Indeed, M. abscessus is resistant to amoebal phagocytosis and the intra-amoeba life seems to increase M. abscessus virulence in an experimental model of infection. However, little is known about M. abscessus virulence in itself. To decipher the genes conferring an advantage to M. abscessus intracellular life, a screening of a M. abscessus transposon mutant library was developed. In parallel, a method of RNA extraction from intracellular Mycobacteria after co-culture with amoebae was developed. This method was validated and allowed the sequencing of whole M. abscessus transcriptomes inside the cells; providing, for the first time, a global view on M. abscessus adaptation to intracellular life. Both approaches give us an insight into M. abscessus virulence factors that enable M. abscessus to colonize the airways in humans.
Collapse
|
18
|
Diop EA, Queiroz EF, Kicka S, Rudaz S, Diop T, Soldati T, Wolfender JL. Survey on medicinal plants traditionally used in Senegal for the treatment of tuberculosis (TB) and assessment of their antimycobacterial activity. JOURNAL OF ETHNOPHARMACOLOGY 2018; 216:71-78. [PMID: 29289797 DOI: 10.1016/j.jep.2017.12.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 12/21/2017] [Accepted: 12/23/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In West Africa, populations are used to taking traditional medicine as a first aid against common health problems. In this aspect, many plants are claimed to be effective in the treatment of Tuberculosis (TB), which according to the World Health Organization (WHO) remains one of the world's deadliest communicable diseases. AIM OF THE STUDY The main aim of this study was to identify plants used to treat TB-symptoms by the population of Senegal and to evaluate their possible concomitant use with clinically approved TB-drugs. This approach allowed the selection of plants effectively used in traditional medicine. In order to verify if the usage of some of these plants can be rationalized, the activity of their traditional preparations was assessed with both an intracellular and extracellular antimycobacterial host-pathogen assays. MATERIALS AND METHODS An ethnopharmacological survey conducted on 117 TB-patients and 30 healers in Senegal from March to May 2014. The questionnaires were focused on the use of medicinal plants to treat common TB -symptoms (cough longer than 2 weeks, fever, night sweats, weight loss and bloody sputum). Local plant names, utilized organs (herbal drugs) and traditional formulations of the plants were recorded. Extracts were prepared by mimicking the traditional decoction in boiling water and screened for their antimycobacterial activity using Mycobacterium marinum, as a validated TB surrogate, and an Acanthamoeba castellanii - M. marinum whole-cell based host-pathogen assay, to detect anti-infective activities. RESULTS By the end of the survey, nearly 30 plants were cited and the 12 most cited herbal drugs were collected and their usage documented by extensive literature search. Extracts of the chosen herbs were screened with the described assays; with a main focus on traditional formulas (mainly herbal decoctions). Two of the water extracts from Combretum aculeatum and Guiera senegalensis showed significant antimycobacterial activities when compared to the positive control drug (rifampin). These extracts showed no observable toxicity against amoeba host cells (Acanthamoeba castellanii). CONCLUSIONS This study demonstrates that most of the patients do not concomitantly use plants and TB drugs (~90% of informants) but, instead, most are treated with medicinal plants before they are admitted to a hospital (41%). Interestingly, among the aqueous extracts assayed, two extracts (Combretum aculeatum (Combretaceae) and Guiera senegalensis (Combretaceae)) collected within this survey demonstrate antimycobacterial activities on the validated whole-cell based host-pathogen assay. Both extracts showed significant activities against intracellular and extracellular - M. marinum growth presenting IC50 lower than 0.5mg/ml compared to the reference drug Rifampin (IC50 of 0.4 and 7µg/ml). No toxicity was observed for amoebae cells at concentration until 0.8mg/ml.
Collapse
Affiliation(s)
- ElHadji Assane Diop
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211 Geneva 11, Switzerland; Biology Department, University Cheikh Anta Diop, Dakar, Senegal
| | - Emerson Ferreira Queiroz
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211 Geneva 11, Switzerland
| | - Sébastien Kicka
- Department of Biochemistry, Faculty of Science, University of Geneva, Quai Ansermet 30, Geneva 4, Switzerland
| | - Serge Rudaz
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211 Geneva 11, Switzerland
| | - Tahir Diop
- Biology Department, University Cheikh Anta Diop, Dakar, Senegal
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Science, University of Geneva, Quai Ansermet 30, Geneva 4, Switzerland
| | - Jean-Luc Wolfender
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211 Geneva 11, Switzerland.
| |
Collapse
|
19
|
Swart AL, Harrison CF, Eichinger L, Steinert M, Hilbi H. Acanthamoeba and Dictyostelium as Cellular Models for Legionella Infection. Front Cell Infect Microbiol 2018; 8:61. [PMID: 29552544 PMCID: PMC5840211 DOI: 10.3389/fcimb.2018.00061] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/13/2018] [Indexed: 12/20/2022] Open
Abstract
Environmental bacteria of the genus Legionella naturally parasitize free-living amoebae. Upon inhalation of bacteria-laden aerosols, the opportunistic pathogens grow intracellularly in alveolar macrophages and can cause a life-threatening pneumonia termed Legionnaires' disease. Intracellular replication in amoebae and macrophages takes place in a unique membrane-bound compartment, the Legionella-containing vacuole (LCV). LCV formation requires the bacterial Icm/Dot type IV secretion system, which translocates literally hundreds of "effector" proteins into host cells, where they modulate crucial cellular processes for the pathogen's benefit. The mechanism of LCV formation appears to be evolutionarily conserved, and therefore, amoebae are not only ecologically significant niches for Legionella spp., but also useful cellular models for eukaryotic phagocytes. In particular, Acanthamoeba castellanii and Dictyostelium discoideum emerged over the last years as versatile and powerful models. Using genetic, biochemical and cell biological approaches, molecular interactions between amoebae and Legionella pneumophila have recently been investigated in detail with a focus on the role of phosphoinositide lipids, small and large GTPases, autophagy components and the retromer complex, as well as on bacterial effectors targeting these host factors.
Collapse
Affiliation(s)
- A Leoni Swart
- Institute of Medical Microbiology, Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Christopher F Harrison
- Max von Pettenkofer Institute, Medical Faculty, Ludwig-Maximilians University Munich, Munich, Germany
| | - Ludwig Eichinger
- Institute for Biochemistry I, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Michael Steinert
- Department of Life Sciences, Institute of Microbiology, Technical University of Braunschweig, Braunschweig, Germany
| | - Hubert Hilbi
- Institute of Medical Microbiology, Medical Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
20
|
Trofimov V, Kicka S, Mucaria S, Hanna N, Ramon-Olayo F, Del Peral LVG, Lelièvre J, Ballell L, Scapozza L, Besra GS, Cox JAG, Soldati T. Antimycobacterial drug discovery using Mycobacteria-infected amoebae identifies anti-infectives and new molecular targets. Sci Rep 2018; 8:3939. [PMID: 29500372 PMCID: PMC5834492 DOI: 10.1038/s41598-018-22228-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 02/06/2018] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis remains a serious threat to human health world-wide, and improved efficiency of medical treatment requires a better understanding of the pathogenesis and the discovery of new drugs. In the present study, we performed a whole-cell based screen in order to complete the characterization of 168 compounds from the GlaxoSmithKline TB-set. We have established and utilized novel previously unexplored host-model systems to characterize the GSK compounds, i.e. the amoeboid organisms D. discoideum and A. castellanii, as well as a microglial phagocytic cell line, BV2. We infected these host cells with Mycobacterium marinum to monitor and characterize the anti-infective activity of the compounds with quantitative fluorescence measurements and high-content microscopy. In summary, 88.1% of the compounds were confirmed as antibiotics against M. marinum, 11.3% and 4.8% displayed strong anti-infective activity in, respectively, the mammalian and protozoan infection models. Additionally, in the two systems, 13–14% of the compounds displayed pro-infective activity. Our studies underline the relevance of using evolutionarily distant pathogen and host models in order to reveal conserved mechanisms of virulence and defence, respectively, which are potential “universal” targets for intervention. Subsequent mechanism of action studies based on generation of over-expresser M. bovis BCG strains, generation of spontaneous resistant mutants and whole genome sequencing revealed four new molecular targets, including FbpA, MurC, MmpL3 and GlpK.
Collapse
Affiliation(s)
- Valentin Trofimov
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland.,Institut Pasteur de Lille, Lille, France
| | - Sébastien Kicka
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Sabrina Mucaria
- Pharmaceutical Biochemistry/Chemistry, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Nabil Hanna
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | | | | | - Joël Lelièvre
- GSK, Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Lluís Ballell
- GSK, Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry/Chemistry, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Gurdyal S Besra
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Jonathan A G Cox
- School of Life & Health Sciences, Aston University, Birmingham, UK.
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
21
|
Cardenal-Muñoz E, Barisch C, Lefrançois LH, López-Jiménez AT, Soldati T. When Dicty Met Myco, a (Not So) Romantic Story about One Amoeba and Its Intracellular Pathogen. Front Cell Infect Microbiol 2018; 7:529. [PMID: 29376033 PMCID: PMC5767268 DOI: 10.3389/fcimb.2017.00529] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/18/2017] [Indexed: 01/06/2023] Open
Abstract
In recent years, Dictyostelium discoideum has become an important model organism to study the cell biology of professional phagocytes. This amoeba not only shares many molecular features with mammalian macrophages, but most of its fundamental signal transduction pathways are conserved in humans. The broad range of existing genetic and biochemical tools, together with its suitability for cell culture and live microscopy, make D. discoideum an ideal and versatile laboratory organism. In this review, we focus on the use of D. discoideum as a phagocyte model for the study of mycobacterial infections, in particular Mycobacterium marinum. We look in detail at the intracellular cycle of M. marinum, from its uptake by D. discoideum to its active or passive egress into the extracellular medium. In addition, we describe the molecular mechanisms that both the mycobacterial invader and the amoeboid host have developed to fight against each other, and compare and contrast with those developed by mammalian phagocytes. Finally, we introduce the methods and specific tools that have been used so far to monitor the D. discoideum-M. marinum interaction.
Collapse
Affiliation(s)
- Elena Cardenal-Muñoz
- Department of Biochemistry, Sciences II, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | | | | | | | | |
Collapse
|
22
|
Dunn JD, Bosmani C, Barisch C, Raykov L, Lefrançois LH, Cardenal-Muñoz E, López-Jiménez AT, Soldati T. Eat Prey, Live: Dictyostelium discoideum As a Model for Cell-Autonomous Defenses. Front Immunol 2018; 8:1906. [PMID: 29354124 PMCID: PMC5758549 DOI: 10.3389/fimmu.2017.01906] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/13/2017] [Indexed: 12/11/2022] Open
Abstract
The soil-dwelling social amoeba Dictyostelium discoideum feeds on bacteria. Each meal is a potential infection because some bacteria have evolved mechanisms to resist predation. To survive such a hostile environment, D. discoideum has in turn evolved efficient antimicrobial responses that are intertwined with phagocytosis and autophagy, its nutrient acquisition pathways. The core machinery and antimicrobial functions of these pathways are conserved in the mononuclear phagocytes of mammals, which mediate the initial, innate-immune response to infection. In this review, we discuss the advantages and relevance of D. discoideum as a model phagocyte to study cell-autonomous defenses. We cover the antimicrobial functions of phagocytosis and autophagy and describe the processes that create a microbicidal phagosome: acidification and delivery of lytic enzymes, generation of reactive oxygen species, and the regulation of Zn2+, Cu2+, and Fe2+ availability. High concentrations of metals poison microbes while metal sequestration inhibits their metabolic activity. We also describe microbial interference with these defenses and highlight observations made first in D. discoideum. Finally, we discuss galectins, TNF receptor-associated factors, tripartite motif-containing proteins, and signal transducers and activators of transcription, microbial restriction factors initially characterized in mammalian phagocytes that have either homologs or functional analogs in D. discoideum.
Collapse
Affiliation(s)
- Joe Dan Dunn
- Faculty of Sciences, Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Cristina Bosmani
- Faculty of Sciences, Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Caroline Barisch
- Faculty of Sciences, Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Lyudmil Raykov
- Faculty of Sciences, Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Louise H Lefrançois
- Faculty of Sciences, Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Elena Cardenal-Muñoz
- Faculty of Sciences, Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | | | - Thierry Soldati
- Faculty of Sciences, Department of Biochemistry, University of Geneva, Geneva, Switzerland
| |
Collapse
|
23
|
Trofimov V, Costa-Gouveia J, Hoffmann E, Brodin P. Host-pathogen systems for early drug discovery against tuberculosis. Curr Opin Microbiol 2017; 39:143-151. [PMID: 29179041 DOI: 10.1016/j.mib.2017.11.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/17/2017] [Indexed: 12/21/2022]
Abstract
Tuberculosis (TB) is a global disease causing 1.8 million deaths each year. The appearance of drug-resistant strains raised the demand for new anti-mycobacterial drugs and therapies, because previously discovered antibiotics are shown to be inefficient. Moreover, the number of newly discovered drugs is not increasing in proportion to the emergence of drug resistance, which suggests that more optimized methodology and screening procedures are required including the incorporation of in vivo properties of TB infection. A way to improve efficacy of screening approaches is by introducing the use of different host-pathogen systems into primary screenings. These include whole cell-based screenings, zebrafish larvae-based screenings and the impact of artificial granuloma research on the drug discovery process. This review highlights current screening attempts and the identified molecular targets and summarizes findings of alternative, not fully explored host-pathogen systems for the characterization of anti-mycobacterial compounds.
Collapse
Affiliation(s)
- Valentin Trofimov
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, University Lille, Lille, France
| | - Joana Costa-Gouveia
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, University Lille, Lille, France
| | - Eik Hoffmann
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, University Lille, Lille, France
| | - Priscille Brodin
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, University Lille, Lille, France.
| |
Collapse
|
24
|
Inhibitors of Mycobacterium marinum virulence identified in a Dictyostelium discoideum host model. PLoS One 2017; 12:e0181121. [PMID: 28727774 PMCID: PMC5519057 DOI: 10.1371/journal.pone.0181121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/26/2017] [Indexed: 12/21/2022] Open
Abstract
Tuberculosis remains one of the major threats to public health worldwide. Given the prevalence of multi drug resistance (MDR) in Mycobacterium tuberculosis strains, there is a strong need to develop new anti-mycobacterial drugs with modes of action distinct from classical antibiotics. Inhibitors of mycobacterial virulence might target new molecular processes and may represent a potential new therapeutic alternative. In this study, we used a Dictyostelium discoideum host model to assess virulence of Mycobacterium marinum and to identify compounds inhibiting mycobacterial virulence. Among 9995 chemical compounds, we selected 12 inhibitors of mycobacterial virulence that do not inhibit mycobacterial growth in synthetic medium. Further analyses revealed that 8 of them perturbed functions requiring an intact mycobacterial cell wall such as sliding motility, bacterial aggregation or cell wall permeability. Chemical analogs of two compounds were analyzed. Chemical modifications altered concomitantly their effect on sliding motility and on mycobacterial virulence, suggesting that the alteration of the mycobacterial cell wall caused the loss of virulence. We characterized further one of the selected compounds and found that it inhibited the ability of mycobacteria to replicate in infected cells. Together these results identify new antimycobacterial compounds that represent new tools to unravel the molecular mechanisms controlling mycobacterial pathogenicity. The isolation of compounds with anti-virulence activity is the first step towards developing new antibacterial treatments.
Collapse
|
25
|
Hochstrasser R, Hilbi H. Intra-Species and Inter-Kingdom Signaling of Legionella pneumophila. Front Microbiol 2017; 8:79. [PMID: 28217110 PMCID: PMC5289986 DOI: 10.3389/fmicb.2017.00079] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/11/2017] [Indexed: 12/24/2022] Open
Abstract
The ubiquitous Gram-negative bacterium Legionella pneumophila parasitizes environ mental amoebae and, upon inhalation, replicates in alveolar macrophages, thus causing a life-threatening pneumonia called “Legionnaires’ disease.” The opportunistic pathogen employs a bi-phasic life cycle, alternating between a replicative, non-virulent phase and a stationary, transmissive/virulent phase. L. pneumophila employs the Lqs (Legionella quorum sensing) system as a major regulator of the growth phase switch. The Lqs system comprises the autoinducer synthase LqsA, the homologous sensor kinases LqsS and LqsT, as well as a prototypic response regulator termed LqsR. These components produce, detect, and respond to the α-hydroxyketone signaling molecule LAI-1 (Legionella autoinducer-1, 3-hydroxypentadecane-4-one). LAI-1-mediated signal transduction through the sensor kinases converges on LqsR, which dimerizes upon phosphorylation. The Lqs system regulates the bacterial growth phase switch, pathogen-host cell interactions, motility, natural competence, filament production, and expression of a chromosomal “fitness island.” Yet, LAI-1 not only mediates bacterial intra-species signaling, but also modulates the motility of eukaryotic cells through the small GTPase Cdc42 and thus promotes inter-kingdom signaling. Taken together, the low molecular weight compound LAI-1 produced by L. pneumophila and sensed by the bacteria as well as by eukaryotic cells plays a major role in pathogen-host cell interactions.
Collapse
Affiliation(s)
- Ramon Hochstrasser
- Department of Medicine, Institute of Medical Microbiology, University of Zürich Zürich, Switzerland
| | - Hubert Hilbi
- Department of Medicine, Institute of Medical Microbiology, University of Zürich Zürich, Switzerland
| |
Collapse
|
26
|
Barisch C, Soldati T. Mycobacterium marinum Degrades Both Triacylglycerols and Phospholipids from Its Dictyostelium Host to Synthesise Its Own Triacylglycerols and Generate Lipid Inclusions. PLoS Pathog 2017; 13:e1006095. [PMID: 28103313 PMCID: PMC5245797 DOI: 10.1371/journal.ppat.1006095] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/28/2016] [Indexed: 12/21/2022] Open
Abstract
During a tuberculosis infection and inside lipid-laden foamy macrophages, fatty acids (FAs) and sterols are the major energy and carbon source for Mycobacterium tuberculosis. Mycobacteria can be found both inside a vacuole and the cytosol, but how this impacts their access to lipids is not well appreciated. Lipid droplets (LDs) store FAs in form of triacylglycerols (TAGs) and are energy reservoirs of prokaryotes and eukaryotes. Using the Dictyostelium discoideum/Mycobacterium marinum infection model we showed that M. marinum accesses host LDs to build up its own intracytosolic lipid inclusions (ILIs). Here, we show that host LDs aggregate at regions of the bacteria that become exposed to the cytosol, and appear to coalesce on their hydrophobic surface leading to a transfer of diacylglycerol O-acyltransferase 2 (Dgat2)-GFP onto the bacteria. Dictyostelium knockout mutants for both Dgat enzymes are unable to generate LDs. Instead, the excess of exogenous FAs is esterified predominantly into phospholipids, inducing uncontrolled proliferation of the endoplasmic reticulum (ER). Strikingly, in absence of host LDs, M. marinum alternatively exploits these phospholipids, resulting in rapid reversal of ER-proliferation. In addition, the bacteria are unable to restrict their acquisition of lipids from the dgat1&2 double knockout leading to vast accumulation of ILIs. Recent data indicate that the presence of ILIs is one of the characteristics of dormant mycobacteria. During Dictyostelium infection, ILI formation in M. marinum is not accompanied by a significant change in intracellular growth and a reduction in metabolic activity, thus providing evidence that storage of neutral lipids does not necessarily induce dormancy.
Collapse
Affiliation(s)
- Caroline Barisch
- Department of Biochemistry, Science II, University of Geneva, 30 quai Ernest-Ansermet, Geneva, Switzerland
- * E-mail:
| | - Thierry Soldati
- Department of Biochemistry, Science II, University of Geneva, 30 quai Ernest-Ansermet, Geneva, Switzerland
| |
Collapse
|
27
|
Mesquita A, Cardenal-Muñoz E, Dominguez E, Muñoz-Braceras S, Nuñez-Corcuera B, Phillips BA, Tábara LC, Xiong Q, Coria R, Eichinger L, Golstein P, King JS, Soldati T, Vincent O, Escalante R. Autophagy in Dictyostelium: Mechanisms, regulation and disease in a simple biomedical model. Autophagy 2016; 13:24-40. [PMID: 27715405 DOI: 10.1080/15548627.2016.1226737] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Autophagy is a fast-moving field with an enormous impact on human health and disease. Understanding the complexity of the mechanism and regulation of this process often benefits from the use of simple experimental models such as the social amoeba Dictyostelium discoideum. Since the publication of the first review describing the potential of D. discoideum in autophagy, significant advances have been made that demonstrate both the experimental advantages and interest in using this model. Since our previous review, research in D. discoideum has shed light on the mechanisms that regulate autophagosome formation and contributed significantly to the study of autophagy-related pathologies. Here, we review these advances, as well as the current techniques to monitor autophagy in D. discoideum. The comprehensive bioinformatics search of autophagic proteins that was a substantial part of the previous review has not been revisited here except for those aspects that challenged previous predictions such as the composition of the Atg1 complex. In recent years our understanding of, and ability to investigate, autophagy in D. discoideum has evolved significantly and will surely enable and accelerate future research using this model.
Collapse
Affiliation(s)
- Ana Mesquita
- a Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid , Spain.,b University of Cincinnati College of Medicine , Cincinnati , OH , USA
| | - Elena Cardenal-Muñoz
- c Départment de Biochimie , Faculté des Sciences, Université de Genève , Switzerland
| | - Eunice Dominguez
- a Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid , Spain.,d Departamento de Genética Molecular , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico City , México
| | - Sandra Muñoz-Braceras
- a Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid , Spain
| | | | - Ben A Phillips
- e Department of Biomedical Sciences , University of Sheffield , UK
| | - Luis C Tábara
- a Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid , Spain
| | - Qiuhong Xiong
- f Center for Biochemistry, Medical Faculty, University of Cologne , Cologne , Germany
| | - Roberto Coria
- d Departamento de Genética Molecular , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico City , México
| | - Ludwig Eichinger
- f Center for Biochemistry, Medical Faculty, University of Cologne , Cologne , Germany
| | - Pierre Golstein
- g Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2 , Inserm, U1104, CNRS UMR7280, Marseille , France
| | - Jason S King
- e Department of Biomedical Sciences , University of Sheffield , UK
| | - Thierry Soldati
- c Départment de Biochimie , Faculté des Sciences, Université de Genève , Switzerland
| | - Olivier Vincent
- a Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid , Spain
| | - Ricardo Escalante
- a Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid , Spain
| |
Collapse
|
28
|
Untargeted Metabolomics To Ascertain Antibiotic Modes of Action. Antimicrob Agents Chemother 2016; 60:2281-91. [PMID: 26833150 PMCID: PMC4808186 DOI: 10.1128/aac.02109-15] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 01/31/2016] [Indexed: 01/23/2023] Open
Abstract
Deciphering the mode of action (MOA) of new antibiotics discovered through phenotypic screening is of increasing importance. Metabolomics offers a potentially rapid and cost-effective means of identifying modes of action of drugs whose effects are mediated through changes in metabolism. Metabolomics techniques also collect data on off-target effects and drug modifications. Here, we present data from an untargeted liquid chromatography-mass spectrometry approach to identify the modes of action of eight compounds: 1-[3-fluoro-4-(5-methyl-2,4-dioxo-pyrimidin-1-yl)phenyl]-3-[2-(trifluoromethyl)phenyl]urea (AZ1), 2-(cyclobutylmethoxy)-5'-deoxyadenosine, triclosan, fosmidomycin, CHIR-090, carbonyl cyanidem-chlorophenylhydrazone (CCCP), 5-chloro-2-(methylsulfonyl)-N-(1,3-thiazol-2-yl)-4-pyrimidinecarboxamide (AZ7), and ceftazidime. Data analysts were blind to the compound identities but managed to identify the target as thymidylate kinase for AZ1, isoprenoid biosynthesis for fosmidomycin, acyl-transferase for CHIR-090, and DNA metabolism for 2-(cyclobutylmethoxy)-5'-deoxyadenosine. Changes to cell wall metabolites were seen in ceftazidime treatments, although other changes, presumably relating to off-target effects, dominated spectral outputs in the untargeted approach. Drugs which do not work through metabolic pathways, such as the proton carrier CCCP, have no discernible impact on the metabolome. The untargeted metabolomics approach also revealed modifications to two compounds, namely, fosmidomycin and AZ7. An untreated control was also analyzed, and changes to the metabolome were seen over 4 h, highlighting the necessity for careful controls in these types of studies. Metabolomics is a useful tool in the analysis of drug modes of action and can complement other technologies already in use.
Collapse
|
29
|
Phagocyte interactions with Mycobacterium tuberculosis--Simultaneous analysis of phagocytosis, phagosome maturation and intracellular replication by imaging flow cytometry. J Immunol Methods 2015; 427:73-84. [PMID: 26476130 DOI: 10.1016/j.jim.2015.10.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/13/2015] [Accepted: 10/13/2015] [Indexed: 02/07/2023]
Abstract
Utilization of compounds that enhance the innate immune response against Mycobacterium tuberculosis is an attractive strategy for combating tuberculosis in the post-antibiotic era. Thus, it is crucial to develop methods that can be used to screen for such compounds and to investigate their mechanisms of action. Here, we used imaging flow cytometry (ImageStreamX Mk II),which enables rapid quantification of microscopic images in flow, to study the interaction between phagocytes and M. tuberculosis. Macrophage-differentiated THP-1 cells were infected with GFP-expressing M. tuberculosis H37Ra, and methods for rapidly assessing phagocytosis, phagosome maturation, and bacterial replication inside the cells were developed and evaluated. These aspects of innate immunity are essential in determining the outcome of mycobacterial infection of phagocytes. The technique was found effective for monitoring phagocytosis of mycobacteria, phagosomal acidification and phagolysosomal fusion, as well as for measuring mycobacterial replication inside the cells. Several of these aspects could be analyzed simultaneously in the same sample, providing a great deal of information about the phagocyte–mycobacterial interaction at once. Thus, this method has great potential to be useful both for basic research questions and for evaluating compounds that enhance the innate immune response against M. tuberculosis.
Collapse
|
30
|
Ang MLT, Murima P, Pethe K. Next-generation antimicrobials: from chemical biology to first-in-class drugs. Arch Pharm Res 2015; 38:1702-17. [PMID: 26259630 PMCID: PMC4567591 DOI: 10.1007/s12272-015-0645-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/29/2015] [Indexed: 01/11/2023]
Abstract
The global emergence of multi-drug resistant bacteria invokes an urgent and imperative necessity for the identification of novel antimicrobials. The general lack of success in progressing novel chemical entities from target-based drug screens have prompted calls for radical and innovative approaches for drug discovery. Recent developments in chemical biology and target deconvolution strategies have revived interests in the utilization of whole-cell phenotypic screens and resulted in several success stories for the discovery and development novel drug candidates and target pathways. In this review, we present and discuss recent chemical biology approaches focusing on the discovery of novel targets and new lead molecules for the treatment of human bacterial and protozoan infections.
Collapse
Affiliation(s)
- Michelle Lay Teng Ang
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, 30 Biopolis Street, #B2-15a, Singapore, 138671, Singapore.
| | - Paul Murima
- Global Health Institute, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Kevin Pethe
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, 30 Biopolis Street, #B2-15a, Singapore, 138671, Singapore.
| |
Collapse
|
31
|
Harrison CF, Chiriano G, Finsel I, Manske C, Hoffmann C, Steiner B, Kranjc A, Patthey-Vuadens O, Kicka S, Trofimov V, Ouertatani-Sakouhi H, Soldati T, Scapozza L, Hilbi H. Amoebae-Based Screening Reveals a Novel Family of Compounds Restricting Intracellular Legionella pneumophila. ACS Infect Dis 2015; 1:327-38. [PMID: 27622823 DOI: 10.1021/acsinfecdis.5b00002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The causative agent of Legionnaires' disease, Legionella pneumophila, grows in environmental amoebae and mammalian macrophages within a distinct compartment, the 'Legionella-containing vacuole' (LCV). Intracellular bacteria are protected from many antibiotics, and thus are notoriously difficult to eradicate. To identify novel compounds that restrict intracellular bacterial replication, we previously developed an assay based on a coculture of amoebae and GFP-producing L. pneumophila. This assay was used to screen a pathway-based, highly diverse chemical library, referred to as the Sinergia library. In this work, we chose to focus on a group of 11 hit compounds, the majority of which originated from the query molecule CN585, a compound that targets the protein phosphatase calcineurin. Further studies on 78 related compound variants revealed crucial structural attributes, namely a triple-ring scaffold with a central triazine moiety, substituted in positions 3 and 5 by two piperidine or pyrrolidine rings, and in position 1 by an amine group bearing a single aliphatic chain moiety. The most effective compound, ZINC00615682, inhibited intracellular replication of L. pneumophila with an IC50 of approximately 20 nM in Acanthamoeba castellanii and slightly less efficiently in Dictyostelium discoideum or macrophages. Pharmacological and genetic attempts to implicate calcineurin in the intracellular replication of L. pneumophila failed. Taken together, these results show that the amoebae-based screen and structure-activity relationship analysis is suitable for the identification of novel inhibitors of the intracellular replication of L. pneumophila. The most potent compound identified in this study targets (an) as yet unidentified host factor(s).
Collapse
Affiliation(s)
- Christopher F. Harrison
- Max von Pettenkofer Institute, Department
of Medicine, Ludwig-Maximilians University Munich, 80336 Munich, Germany
| | - Gianpaolo Chiriano
- School of Pharmaceutical
Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 1211 Geneva, Switzerland
| | - Ivo Finsel
- Max von Pettenkofer Institute, Department
of Medicine, Ludwig-Maximilians University Munich, 80336 Munich, Germany
| | - Christian Manske
- Max von Pettenkofer Institute, Department
of Medicine, Ludwig-Maximilians University Munich, 80336 Munich, Germany
| | - Christine Hoffmann
- Max von Pettenkofer Institute, Department
of Medicine, Ludwig-Maximilians University Munich, 80336 Munich, Germany
| | - Bernhard Steiner
- Institute of Medical Microbiology, Department of Medicine, University of Zurich, Gloriastrasse 30/32, 8006 Zurich, Switzerland
| | - Agata Kranjc
- School of Pharmaceutical
Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 1211 Geneva, Switzerland
| | - Ophelie Patthey-Vuadens
- School of Pharmaceutical
Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 1211 Geneva, Switzerland
| | | | | | | | | | - Leonardo Scapozza
- School of Pharmaceutical
Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 1211 Geneva, Switzerland
| | - Hubert Hilbi
- Max von Pettenkofer Institute, Department
of Medicine, Ludwig-Maximilians University Munich, 80336 Munich, Germany
- Institute of Medical Microbiology, Department of Medicine, University of Zurich, Gloriastrasse 30/32, 8006 Zurich, Switzerland
| |
Collapse
|
32
|
Harrison CF, Kicka S, Kranjc A, Finsel I, Chiriano G, Ouertatani-Sakouhi H, Soldati T, Scapozza L, Hilbi H. Adrenergic antagonists restrict replication of Legionella. MICROBIOLOGY-SGM 2015; 161:1392-406. [PMID: 25873585 DOI: 10.1099/mic.0.000094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Legionella pneumophila is a facultative intracellular bacterium, which upon inhalation can cause a potentially fatal pneumonia termed Legionnaires' disease. The opportunistic pathogen grows in environmental amoebae and mammalian macrophages within a unique membrane-bound compartment, the 'Legionella-containing vacuole'. Bacteria are exposed to many environmental cues including small signalling molecules from eukaryotic cells. A number of pathogenic bacteria sense and respond to catecholamine hormones, such as adrenalin and noradrenalin, a process mediated via the QseBC two-component system in some bacteria. In this study, we examined the effect of adrenergic compounds on L. pneumophila, and discovered that the adrenergic receptor antagonists benoxathian, naftopidil, propranolol and labetalol, as well as the QseC sensor kinase inhibitor LED209, reduced the growth of L. pneumophila in broth or amoebae, while replication in macrophages was enhanced. Growth restriction was common to members of the genus Legionella and Mycobacterium, and was observed for L. pneumophila in the replicative but not stationary phase of the biphasic life cycle. Deletion of the L. pneumophila qseBC genes indicated that growth inhibition by adrenergics or LED209 is mediated only to a minor extent by this two-component system, implying the presence of other adrenergic sensing systems. This study identifies adrenergic molecules as novel inhibitors of extra- and intracellular growth of Legionella and reveals LED209 as a potential lead compound to combat infections with Legionella or Mycobacterium spp.
Collapse
Affiliation(s)
- Christopher F Harrison
- 1Max von Pettenkofer Institute, Department of Medicine, Ludwig-Maximilians University Munich, 80336 Munich, Germany
| | - Sébastien Kicka
- 2Department of Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Agata Kranjc
- 3School of Pharmaceutical Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 1211 Geneva, Switzerland
| | - Ivo Finsel
- 1Max von Pettenkofer Institute, Department of Medicine, Ludwig-Maximilians University Munich, 80336 Munich, Germany
| | - Gianpaolo Chiriano
- 3School of Pharmaceutical Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 1211 Geneva, Switzerland
| | | | - Thierry Soldati
- 2Department of Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Leonardo Scapozza
- 3School of Pharmaceutical Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 1211 Geneva, Switzerland
| | - Hubert Hilbi
- 1Max von Pettenkofer Institute, Department of Medicine, Ludwig-Maximilians University Munich, 80336 Munich, Germany 5Institute of Medical Microbiology, Department of Medicine, University of Zurich, Gloriastrasse 30/32, 8006 Zurich, Switzerland
| |
Collapse
|
33
|
Drancourt M. Looking in amoebae as a source of mycobacteria. Microb Pathog 2014; 77:119-24. [PMID: 25017516 DOI: 10.1016/j.micpath.2014.07.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/01/2014] [Accepted: 07/03/2014] [Indexed: 11/16/2022]
Abstract
Mycobacteria exhibit various relationships with amoebae, ranging from the killing of one partner by the other one, to amoebae hosting mycobacteria in trophozoites and cysts. This observation indicates that poorly described biological factors affect the relationships, including mycobacterial cell-wall glycolipids and the size of the mycobacteria. Experimental observations indicate that a majority of environmental, opportunistic mycobacteria but also obligate pathogens including Mycobacterium tuberculosis, Mycobacterium leprae and Mycobacterium ulcerans are inter-amoebal organisms. Amoebae may give opportunities for genetic exchanges between mycobacteria, sympatric intra-amoebal organisms and the amoebae themselves. Amoebae clearly protect opportunistic mycobacterial pathogens during their environmental life but their role for obligate mycobacterial infection remains to be established. Accordingly, water was the source for emerging, community-acquired and health care-associated infection with amoeba-resisting mycobacteria of the Mycobacterium avium, Mycobacterium abscessus and Mycobacterium fortuitum groups, among others. Amoebae are organisms where mycobacteria can be found and, accordingly, amoeba co-culture can be used for the isolation of mycobacteria from environmental and clinical specimens. Looking in amoebae may help recovering new species of mycobacteria.
Collapse
Affiliation(s)
- M Drancourt
- Aix Marseille Université, URMITE, UM 63 UMR_S1095 UMR 7278, Méditerranée Infection, 13385, Marseille, France.
| |
Collapse
|