1
|
Vidal M, Arch M, Fuentes E, Cardona PJ. Drosophila melanogaster experimental model to test new antimicrobials: a methodological approach. Front Microbiol 2024; 15:1478263. [PMID: 39568995 PMCID: PMC11576456 DOI: 10.3389/fmicb.2024.1478263] [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: 08/09/2024] [Accepted: 10/24/2024] [Indexed: 11/22/2024] Open
Abstract
Given the increasing concern about antimicrobial resistance among the microorganisms that cause infections in our society, there is an urgent need for new drug discovery. Currently, this process involves testing many low-quality compounds, resulting from the in vivo testing, on mammal models, which not only wastes time, resources, and money, but also raises ethical questions. In this review, we have discussed the potential of D. melanogaster as an intermediary experimental model in this drug discovery timeline. We have tackled the topic from a methodological perspective, providing recommendations regarding the range of drug concentrations to test based on the mechanism of action of each compound; how to treat D. melanogaster, how to monitor that treatment, and what parameters we should consider when designing a drug screening protocol to maximize the study's benefits. We also discuss the necessary improvements needed to establish the D. melanogaster model of infection as a standard technique in the drug screening process. Overall, D. melanogaster has been demonstrated to be a manageable model for studying broad-spectrum infection treatment. It allows us to obtain valuable information in a cost-effective manner, which can improve the drug screening process and provide insights into our current major concern. This approach is also in line with the 3R policy in biomedical research, in particular on the replacement and reduce the use of vertebrates in preclinical development.
Collapse
Affiliation(s)
- Maria Vidal
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Marta Arch
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Esther Fuentes
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Pere-Joan Cardona
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
2
|
Longo BM, Trunfio M, Calcagno A. Dual β-lactams for the treatment of Mycobacterium abscessus: a review of the evidence and a call to act against an antibiotic nightmare. J Antimicrob Chemother 2024; 79:2731-2741. [PMID: 39150384 PMCID: PMC11932079 DOI: 10.1093/jac/dkae288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024] Open
Abstract
Mycobacterium abscessus complex is a group of rapidly growing non-tuberculous mycobacteria (NTM), increasingly emerging as opportunistic pathogens. Current treatment options for these microorganisms are limited and associated with a high rate of treatment failure, toxicity and recurrence. In search of new therapeutic strategies, interest has grown in dual β-lactam (DBL) therapy, as research recently discovered that M. abscessus cell wall synthesis is mainly regulated by two types of enzymes (d,d-transpeptidases and l,d-transpeptidases) differently susceptible to inhibition by distinct β-lactams. In vitro studies testing several DBL combinations have shown synergy in extracellular broth cultures as well as in the intracellular setting: cefoxitin/imipenem, ceftaroline/imipenem, ceftazidime/ceftaroline and ceftazidime/imipenem. The addition of specific β-lactamase inhibitors (BLIs) targeting M. abscessus β-lactamase did not significantly enhance the activity of DBL combinations. However, in vivo data are lacking. We reviewed the literature on DBL/DBL-BLI-based therapies for M. abscessus infections to raise greater attention on this promising yet overlooked treatment option and to guide future preclinical and clinical studies.
Collapse
Affiliation(s)
- Bianca Maria Longo
- Department of Medical Sciences, Unit of Infectious Diseases, Amedeo di Savoia Hospital, University of Turin, 10149 Turin, Italy
| | - Mattia Trunfio
- Department of Medical Sciences, Unit of Infectious Diseases, Amedeo di Savoia Hospital, University of Turin, 10149 Turin, Italy
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, CA 92037, USA
| | - Andrea Calcagno
- Department of Medical Sciences, Unit of Infectious Diseases, Amedeo di Savoia Hospital, University of Turin, 10149 Turin, Italy
| |
Collapse
|
3
|
Lagune M, Kremer L, Herrmann JL. Mycobacterium abscessus, a complex of three fast-growing subspecies sharing virulence traits with slow-growing mycobacteria. Clin Microbiol Infect 2024; 30:726-731. [PMID: 37797823 DOI: 10.1016/j.cmi.2023.08.036] [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: 05/15/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND Mycobacterium abscessus belongs to the largest group of mycobacteria, the rapid-growing saprophytic mycobacteria, and is one of the most difficult-to-treat opportunistic pathogen. Several features pertain to the high adaptability of M. abscessus to the host. These include the capacity to survive and persist within amoebae, to transition from a smooth to a rough morphotype that occurs during the course of the disease and to express of a wide array of virulence factors. OBJECTIVES The main objective of this narrative review consists to report major assets of M. abscessus that contribute to the virulence of these rapid-growing saprophytic mycobacteria. Strikingly, many of these determinants, whether they are from a mycobacterial origin or acquired by horizontal gene transfer, are known virulence factors found in slow-growing and strict pathogens for humans and animals. SOURCES In the light of recent published work in the field we attempted to highlight major features characterizing M. abscessus pathogenicity and to explain why this led to the emergence of this mycobacterial species in patients with cystic fibrosis. CONTENT M. abscessus genome plasticity, the smooth-to-rough transition, and the expression of a panel of enzymes associated with virulence in other bacteria are key players in M. abscessus virulence. In addition, the very large repertoire of lipid transporters, known as mycobacterial membrane protein large and small (MmpL and MmpS respectively), deeply influences the pathogenicity of M. abscessus, as exemplified here for some of them. IMPLICATIONS All these traits largely contribute to make M. abscessus a unique mycobacterium regarding to its pathophysiological processes, ranging from the early colonization steps to the establishment of severe and chronic pulmonary diseases.
Collapse
Affiliation(s)
- Marion Lagune
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France; INSERM, IRIM, Montpellier, France
| | - Jean-Louis Herrmann
- Université Paris-Saclay, UVSQ, INSERM, U1173 Infection et Inflammation, Montigny-le-Bretonneux, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Ile-de-France Ouest, GHU Paris-Saclay, Hôpital Raymond Poincaré, Garches, France.
| |
Collapse
|
4
|
Touré H, Herrmann JL, Szuplewski S, Girard-Misguich F. Drosophila melanogaster as an organism model for studying cystic fibrosis and its major associated microbial infections. Infect Immun 2023; 91:e0024023. [PMID: 37847031 PMCID: PMC10652941 DOI: 10.1128/iai.00240-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023] Open
Abstract
Cystic fibrosis (CF) is a human genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator gene that encodes a chloride channel. The most severe clinical manifestation is associated with chronic pulmonary infections by pathogenic and opportunistic microbes. Drosophila melanogaster has become the invertebrate model of choice for modeling microbial infections and studying the induced innate immune response. Here, we review its contribution to the understanding of infections with six major pathogens associated with CF (Staphylococcus aureus, Pseudomonas aeruginosa, Burkholderia cepacia, Mycobacterium abscessus, Streptococcus pneumoniae, and Aspergillus fumigatus) together with the perspectives opened by the recent availability of two CF models in this model organism.
Collapse
Affiliation(s)
- Hamadoun Touré
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-le-Bretonneux, France
| | - Jean-Louis Herrmann
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-le-Bretonneux, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Ile-de-France Ouest, GHU Paris-Saclay, Hôpital Raymond Poincaré, Garches, France
| | - Sébastien Szuplewski
- Université Paris-Saclay, UVSQ, Laboratoire de Génétique et Biologie Cellulaire, Montigny-le-Bretonneux, France
| | - Fabienne Girard-Misguich
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-le-Bretonneux, France
| |
Collapse
|
5
|
Admella J, Torrents E. Investigating bacterial infections in Galleria mellonella larvae: Insights into pathogen dissemination and behavior. J Invertebr Pathol 2023; 200:107975. [PMID: 37541571 DOI: 10.1016/j.jip.2023.107975] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/04/2023] [Accepted: 07/30/2023] [Indexed: 08/06/2023]
Abstract
The insect Galleria mellonella is an alternative animal model widely used for studying bacterial infections. It presents a wide range of advantages, including its low cost, easy maintenance and lack of ethical constraints. Among other features, their innate immune system is very similar to that of mammals. In this study, we dissected several larvae infected with important human pathogens: Mycobacterium abscessus, Staphylococcus aureus and Pseudomonas aeruginosa. By observing the fat body, gut, trachea, and hemolymph under the microscope, we were able to describe where bacteria tend to disseminate. We also quantified the number of bacteria in the hemolymph throughout the infection course and found significant differences between the different pathogens. With this work, we aimed to better understand the behavior and dissemination of bacteria in the infected larvae.
Collapse
Affiliation(s)
- Joana Admella
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15-21, 08028 Barcelona, Spain; Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, 643 Diagonal Ave., 08028 Barcelona, Spain
| | - Eduard Torrents
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15-21, 08028 Barcelona, Spain; Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, 643 Diagonal Ave., 08028 Barcelona, Spain.
| |
Collapse
|
6
|
Touré H, Durand N, Guénal I, Herrmann JL, Girard-Misguich F, Szuplewski S. Mycobacterium abscessus Opsonization Allows an Escape from the Defensin Bactericidal Action in Drosophila. Microbiol Spectr 2023; 11:e0077723. [PMID: 37260399 PMCID: PMC10434004 DOI: 10.1128/spectrum.00777-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/05/2023] [Indexed: 06/02/2023] Open
Abstract
Mycobacterium abscessus, an intracellular nontuberculous mycobacterium, is considered the most pathogenic species among the group of rapidly growing mycobacteria. The resistance of M. abscessus to the host innate response contributes to its pathogenicity in addition to several virulence factors. We have recently shown in Drosophila that antimicrobial peptides (AMPs), whose production is induced by M. abscessus, are unable to control mycobacterial infection. This could be due to their inability to kill mycobacteria and/or the hidden location of the pathogen in phagocytic cells. Here, we demonstrate that the rapid internalization of M. abscessus by Drosophila macrophages allows it to escape the AMP-mediated humoral response. By depleting phagocytes in AMP-deficient flies, we found that several AMPs were required for the control of extracellular M. abscessus. This was confirmed in the Tep4 opsonin-deficient flies, which we show can better control M. abscessus growth and have increased survival through overproduction of some AMPs, including Defensin. Furthermore, Defensin alone was sufficient to kill extracellular M. abscessus both in vitro and in vivo and control its infection. Collectively, our data support that Tep4-mediated opsonization of M. abscessus allows its escape and resistance toward the Defensin bactericidal action in Drosophila. IMPORTANCE Mycobacterium abscessus, an opportunistic pathogen in cystic fibrosis patients, is the most pathogenic species among the fast-growing mycobacteria. How M. abscessus resists the host innate response before establishing an infection remains unclear. Using Drosophila, we have recently demonstrated that M. abscessus resists the host innate response by surviving the cytotoxic lysis of the infected phagocytes and the induced antimicrobial peptides (AMPs), including Defensin. In this work, we demonstrate that M. abscessus resists the latter response by being rapidly internalized by Drosophila phagocytes. Indeed, by combining in vivo and in vitro approaches, we show that Defensin is able to control extracellular M. abscessus infection through a direct bactericidal action. In conclusion, we report that M. abscessus escapes the host AMP-mediated humoral response by taking advantage of its internalization by the phagocytes.
Collapse
Affiliation(s)
- Hamadoun Touré
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
| | - Nicolas Durand
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
| | | | - Jean-Louis Herrmann
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Ile-de-France Ouest, GHU Paris-Saclay, Hôpital Raymond Poincaré, Garches, France
| | - Fabienne Girard-Misguich
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
| | | |
Collapse
|
7
|
Touré H, Galindo LA, Lagune M, Glatigny S, Waterhouse RM, Guénal I, Herrmann JL, Girard-Misguich F, Szuplewski S. Mycobacterium abscessus resists the innate cellular response by surviving cell lysis of infected phagocytes. PLoS Pathog 2023; 19:e1011257. [PMID: 36972320 PMCID: PMC10079227 DOI: 10.1371/journal.ppat.1011257] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 04/06/2023] [Accepted: 02/28/2023] [Indexed: 03/29/2023] Open
Abstract
Mycobacterium abscessus is the most pathogenic species among the predominantly saprophytic fast-growing mycobacteria. This opportunistic human pathogen causes severe infections that are difficult to eradicate. Its ability to survive within the host was described mainly with the rough (R) form of M. abscessus, which is lethal in several animal models. This R form is not present at the very beginning of the disease but appears during the progression and the exacerbation of the mycobacterial infection, by transition from a smooth (S) form. However, we do not know how the S form of M. abscessus colonizes and infects the host to then multiply and cause the disease. In this work, we were able to show the hypersensitivity of fruit flies, Drosophila melanogaster, to intrathoracic infections by the S and R forms of M. abscessus. This allowed us to unravel how the S form resists the innate immune response developed by the fly, both the antimicrobial peptides- and cellular-dependent immune responses. We demonstrate that intracellular M. abscessus was not killed within the infected phagocytic cells, by resisting lysis and caspase-dependent apoptotic cell death of Drosophila infected phagocytes. In mice, in a similar manner, intra-macrophage M. abscessus was not killed when M. abscessus-infected macrophages were lysed by autologous natural killer cells. These results demonstrate the propensity of the S form of M. abscessus to resist the host’s innate responses to colonize and multiply within the host.
Collapse
Affiliation(s)
- Hamadoun Touré
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
| | - Lee Ann Galindo
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
| | - Marion Lagune
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
| | - Simon Glatigny
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
| | - Robert M. Waterhouse
- Department of Ecology and Evolution, University of Lausanne, and the Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Jean-Louis Herrmann
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Ile-de-France Ouest, GHU Paris-Saclay, Hôpital Raymond Poincaré, Garches, France
| | - Fabienne Girard-Misguich
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
- * E-mail: (FGM); (SS)
| | | |
Collapse
|
8
|
Asai M, Li Y, Newton SM, Robertson BD, Langford PR. Galleria mellonella-intracellular bacteria pathogen infection models: the ins and outs. FEMS Microbiol Rev 2023; 47:fuad011. [PMID: 36906279 PMCID: PMC10045907 DOI: 10.1093/femsre/fuad011] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/13/2023] Open
Abstract
Galleria mellonella (greater wax moth) larvae are used widely as surrogate infectious disease models, due to ease of use and the presence of an innate immune system functionally similar to that of vertebrates. Here, we review G. mellonella-human intracellular bacteria pathogen infection models from the genera Burkholderia, Coxiella, Francisella, Listeria, and Mycobacterium. For all genera, G. mellonella use has increased understanding of host-bacterial interactive biology, particularly through studies comparing the virulence of closely related species and/or wild-type versus mutant pairs. In many cases, virulence in G. mellonella mirrors that found in mammalian infection models, although it is unclear whether the pathogenic mechanisms are the same. The use of G. mellonella larvae has speeded up in vivo efficacy and toxicity testing of novel antimicrobials to treat infections caused by intracellular bacteria: an area that will expand since the FDA no longer requires animal testing for licensure. Further use of G. mellonella-intracellular bacteria infection models will be driven by advances in G. mellonella genetics, imaging, metabolomics, proteomics, and transcriptomic methodologies, alongside the development and accessibility of reagents to quantify immune markers, all of which will be underpinned by a fully annotated genome.
Collapse
Affiliation(s)
- Masanori Asai
- Section of Paediatric Infectious Disease, Department of Infectious Disease, St Mary’s campus, Imperial College London, London W2 1PG, United Kingdom
| | - Yanwen Li
- Section of Paediatric Infectious Disease, Department of Infectious Disease, St Mary’s campus, Imperial College London, London W2 1PG, United Kingdom
| | - Sandra M Newton
- Section of Paediatric Infectious Disease, Department of Infectious Disease, St Mary’s campus, Imperial College London, London W2 1PG, United Kingdom
| | - Brian D Robertson
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, South Kensington campus, Imperial College London, London SW7 2AZ, United Kingdom
| | - Paul R Langford
- Section of Paediatric Infectious Disease, Department of Infectious Disease, St Mary’s campus, Imperial College London, London W2 1PG, United Kingdom
| |
Collapse
|
9
|
Sullivan MR, McGowen K, Liu Q, Akusobi C, Young DC, Mayfield JA, Raman S, Wolf ID, Moody DB, Aldrich CC, Muir A, Rubin EJ. Biotin-dependent cell envelope remodelling is required for Mycobacterium abscessus survival in lung infection. Nat Microbiol 2023; 8:481-497. [PMID: 36658396 PMCID: PMC9992005 DOI: 10.1038/s41564-022-01307-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 12/14/2022] [Indexed: 01/21/2023]
Abstract
Mycobacterium abscessus is an emerging pathogen causing lung infection predominantly in patients with underlying structural abnormalities or lung disease and is resistant to most frontline antibiotics. As the pathogenic mechanisms of M. abscessus in the context of the lung are not well-understood, we developed an infection model using air-liquid interface culture and performed a transposon mutagenesis and sequencing screen to identify genes differentially required for bacterial survival in the lung. Biotin cofactor synthesis was required for M. abscessus growth due to increased intracellular biotin demand, while pharmacological inhibition of biotin synthesis prevented bacterial proliferation. Biotin was required for fatty acid remodelling, which increased cell envelope fluidity and promoted M. abscessus survival in the alkaline lung environment. Together, these results indicate that biotin-dependent fatty acid remodelling plays a critical role in pathogenic adaptation to the lung niche, suggesting that biotin synthesis and fatty acid metabolism might provide therapeutic targets for treatment of M. abscessus infection.
Collapse
Affiliation(s)
- Mark R Sullivan
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kerry McGowen
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Qiang Liu
- Department of Medicinal Chemistry, University of Minnesota College of Pharmacy, Minneapolis, MN, USA
| | - Chidiebere Akusobi
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - David C Young
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jacob A Mayfield
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sahadevan Raman
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ian D Wolf
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - D Branch Moody
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Courtney C Aldrich
- Department of Medicinal Chemistry, University of Minnesota College of Pharmacy, Minneapolis, MN, USA
| | - Alexander Muir
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Eric J Rubin
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
10
|
Updated Review on the Mechanisms of Pathogenicity in Mycobacterium abscessus, a Rapidly Growing Emerging Pathogen. Microorganisms 2022; 11:microorganisms11010090. [PMID: 36677382 PMCID: PMC9866562 DOI: 10.3390/microorganisms11010090] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 01/01/2023] Open
Abstract
In recent years, Mycobacterium abscessus has appeared as an emerging pathogen, with an increasing number of disease cases reported worldwide that mainly occur among patients with chronic lung diseases or impaired immune systems. The treatment of this pathogen represents a challenge due to the multi-drug-resistant nature of this species and its ability to evade most therapeutic approaches. However, although predisposing host factors for disease are well known, intrinsic pathogenicity mechanisms of this mycobacterium are still not elucidated. Like other mycobacteria, intracellular invasiveness and survival inside different cell lines are pathogenic factors related to the ability of M. abscessus to establish infection. Some of the molecular factors involved in this process are well-known and are present in the mycobacterial cell wall, such as trehalose-dimycolate and glycopeptidolipids. The ability to form biofilms is another pathogenic factor that is essential for the development of chronic disease and for promoting mycobacterial survival against the host immune system or different antibacterial treatments. This capability also seems to be related to glycopeptidolipids and other lipid molecules, and some studies have shown an intrinsic relationship between both pathogenic mechanisms. Antimicrobial resistance is also considered a mechanism of pathogenicity because it allows the mycobacterium to resist antimicrobial therapies and represents an advantage in polymicrobial biofilms. The recent description of hyperpathogenic strains with the potential interhuman transmission makes it necessary to increase our knowledge of pathogenic mechanisms of this species to design better therapeutic approaches to the management of these infections.
Collapse
|
11
|
Boeck L, Burbaud S, Skwark M, Pearson WH, Sangen J, Wuest AW, Marshall EKP, Weimann A, Everall I, Bryant JM, Malhotra S, Bannerman BP, Kierdorf K, Blundell TL, Dionne MS, Parkhill J, Andres Floto R. Mycobacterium abscessus pathogenesis identified by phenogenomic analyses. Nat Microbiol 2022; 7:1431-1441. [PMID: 36008617 PMCID: PMC9418003 DOI: 10.1038/s41564-022-01204-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/19/2022] [Indexed: 12/12/2022]
Abstract
The medical and scientific response to emerging and established pathogens is often severely hampered by ignorance of the genetic determinants of virulence, drug resistance and clinical outcomes that could be used to identify therapeutic drug targets and forecast patient trajectories. Taking the newly emergent multidrug-resistant bacteria Mycobacterium abscessus as an example, we show that combining high-dimensional phenotyping with whole-genome sequencing in a phenogenomic analysis can rapidly reveal actionable systems-level insights into bacterial pathobiology. Through phenotyping of 331 clinical isolates, we discovered three distinct clusters of isolates, each with different virulence traits and associated with a different clinical outcome. We combined genome-wide association studies with proteome-wide computational structural modelling to define likely causal variants, and employed direct coupling analysis to identify co-evolving, and therefore potentially epistatic, gene networks. We then used in vivo CRISPR-based silencing to validate our findings and discover clinically relevant M. abscessus virulence factors including a secretion system, thus illustrating how phenogenomics can reveal critical pathways within emerging pathogenic bacteria.
Collapse
Affiliation(s)
- Lucas Boeck
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK
- Cambridge Centre for AI in Medicine, Cambridge, UK
- Wellcome Sanger Institute, Hinxton, UK
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Sophie Burbaud
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK
- Cambridge Centre for AI in Medicine, Cambridge, UK
| | - Marcin Skwark
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Will H Pearson
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
- Department of Life Sciences, Imperial College London, London, UK
| | - Jasper Sangen
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK
- Cambridge Centre for AI in Medicine, Cambridge, UK
| | - Andreas W Wuest
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Eleanor K P Marshall
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
- Department of Life Sciences, Imperial College London, London, UK
| | - Aaron Weimann
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK
- Cambridge Centre for AI in Medicine, Cambridge, UK
| | | | - Josephine M Bryant
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK
- Cambridge Centre for AI in Medicine, Cambridge, UK
| | - Sony Malhotra
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Scientific Computing Department, Science and Technology Facilities Council, Harwell, UK
| | - Bridget P Bannerman
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK
- Cambridge Centre for AI in Medicine, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Katrin Kierdorf
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
- Department of Life Sciences, Imperial College London, London, UK
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Marc S Dionne
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
- Department of Life Sciences, Imperial College London, London, UK
| | - Julian Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - R Andres Floto
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK.
- Cambridge Centre for AI in Medicine, Cambridge, UK.
- Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, UK.
| |
Collapse
|
12
|
Abdelaal HFM, Chan ED, Young L, Baldwin SL, Coler RN. Mycobacterium abscessus: It's Complex. Microorganisms 2022; 10:1454. [PMID: 35889173 PMCID: PMC9316637 DOI: 10.3390/microorganisms10071454] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 12/21/2022] Open
Abstract
Mycobacterium abscessus (M. abscessus) is an opportunistic pathogen usually colonizing abnormal lung airways and is often seen in patients with cystic fibrosis. Currently, there is no vaccine available for M. abscessus in clinical development. The treatment of M. abscessus-related pulmonary diseases is peculiar due to intrinsic resistance to several commonly used antibiotics. The development of either prophylactic or therapeutic interventions for M. abscessus pulmonary infections is hindered by the absence of an adequate experimental animal model. In this review, we outline the critical elements related to M. abscessus virulence mechanisms, host-pathogen interactions, and treatment challenges associated with M. abscessus pulmonary infections. The challenges of effectively combating this pathogen include developing appropriate preclinical animal models of infection, developing proper diagnostics, and designing novel strategies for treating drug-resistant M. abscessus.
Collapse
Affiliation(s)
- Hazem F. M. Abdelaal
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98145, USA; (H.F.M.A.); (S.L.B.)
| | - Edward D. Chan
- Department of Academic Affairs and Medicine, National Jewish Health, Denver, CO 80206, USA;
- Pulmonary Section, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045, USA
| | - Lisa Young
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
| | - Susan L. Baldwin
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98145, USA; (H.F.M.A.); (S.L.B.)
| | - Rhea N. Coler
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98145, USA; (H.F.M.A.); (S.L.B.)
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
13
|
Kam JY, Hortle E, Krogman E, Warner SE, Wright K, Luo K, Cheng T, Manuneedhi Cholan P, Kikuchi K, Triccas JA, Britton WJ, Johansen MD, Kremer L, Oehlers SH. Rough and smooth variants of Mycobacterium abscessus are differentially controlled by host immunity during chronic infection of adult zebrafish. Nat Commun 2022; 13:952. [PMID: 35177649 PMCID: PMC8854618 DOI: 10.1038/s41467-022-28638-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 02/03/2022] [Indexed: 11/09/2022] Open
Abstract
Prevalence of Mycobacterium abscessus infections is increasing in patients with respiratory comorbidities. After initial colonisation, M. abscessus smooth colony (S) variants can undergo an irreversible genetic switch into highly inflammatory, rough colony (R) variants, often associated with a decline in pulmonary function. Here, we use an adult zebrafish model of chronic infection with R and S variants to study M. abscessus pathogenesis in the context of fully functioning host immunity. We show that infection with an R variant causes an inflammatory immune response that drives necrotic granuloma formation through host TNF signalling, mediated by the tnfa, tnfr1 and tnfr2 gene products. T cell-dependent immunity is stronger against the R variant early in infection, and regulatory T cells associate with R variant granulomas and limit bacterial growth. In comparison, an S variant proliferates to high burdens but appears to be controlled by TNF-dependent innate immunity early during infection, resulting in delayed granuloma formation. Thus, our work demonstrates the applicability of adult zebrafish to model persistent M. abscessus infection, and illustrates differences in the immunopathogenesis induced by R and S variants during granulomatous infection. The pathogen Mycobacterium abscessus can switch from a smooth colony form (S) into a highly inflammatory, rough colony form (R) during infection. Here, Kam et al. use an adult zebrafish model of M. abscessus chronic infection to illustrate differences in the immunopathogenesis induced by R and S variants.
Collapse
Affiliation(s)
- Julia Y Kam
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Elinor Hortle
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.,The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, NSW, Australia
| | - Elizabeth Krogman
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Sherridan E Warner
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.,The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, NSW, Australia
| | - Kathryn Wright
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Kaiming Luo
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Tina Cheng
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Pradeep Manuneedhi Cholan
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Kazu Kikuchi
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Kensington, NSW, Australia
| | - James A Triccas
- The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, NSW, Australia
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.,Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Matt D Johansen
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France.,INSERM, IRIM, Montpellier, France
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia. .,The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, NSW, Australia. .,A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
14
|
Marshall EKP, Dionne MS. Drosophila versus Mycobacteria: A model for mycobacterial host-pathogen interactions. Mol Microbiol 2021; 117:600-609. [PMID: 34585797 DOI: 10.1111/mmi.14819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 02/01/2023]
Abstract
Animal models have played an essential role in understanding the host-pathogen interactions of pathogenic mycobacteria, including the Mycobacterium tuberculosis and emerging nontuberculous mycobacteria (NTM) species such as M. avium and M. abscessus. Drosophila melanogaster has become a well-established model for the study of innate immunity and is increasingly being used as a tool to study host-pathogen interactions, in part due to its genetic tractability. The use of D. melanogaster has led to greater understanding of the role of the innate immune system in response to mycobacterial infection, including in vitro RNAi screens and in vivo studies. These studies have identified processes and host factors involved in mycobacterial infection, such as those required for cellular entry, those required to control or resist non-pathogenic mycobacteria, or factors that become dysregulated as a result of mycobacterial infection. Developments in genetic tools for manipulating mycobacterial genomes will allow for more detailed studies into how specific host and pathogen factors interact with one another by using D. melanogaster; however, the full potential of this model has not yet been reached. Here we provide an overview of how D. melanogaster has been used to study mycobacterial infection and discuss the current gaps in our understanding.
Collapse
Affiliation(s)
- Eleanor K P Marshall
- Department of Life Sciences, Imperial College London, London, UK.,MRC Centre for Molecular Bacteriology and infection, Imperial College London, London, UK
| | - Marc S Dionne
- Department of Life Sciences, Imperial College London, London, UK.,MRC Centre for Molecular Bacteriology and infection, Imperial College London, London, UK
| |
Collapse
|
15
|
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
|
16
|
Younes S, Al-Sulaiti A, Nasser EAA, Najjar H, Kamareddine L. Drosophila as a Model Organism in Host-Pathogen Interaction Studies. Front Cell Infect Microbiol 2020; 10:214. [PMID: 32656090 PMCID: PMC7324642 DOI: 10.3389/fcimb.2020.00214] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/20/2020] [Indexed: 12/29/2022] Open
Abstract
Owing to the genetic similarities and conserved pathways between a fruit fly and mammals, the use of the Drosophila model as a platform to unveil novel mechanisms of infection and disease progression has been justified and widely instigated. Gaining proper insight into host-pathogen interactions and identifying chief factors involved in host defense and pathogen virulence in Drosophila serves as a foundation to establish novel strategies for infectious disease prevention and control in higher organisms, including humans.
Collapse
Affiliation(s)
- Salma Younes
- Biomedical Sciences Department, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Asma Al-Sulaiti
- Biomedical Sciences Department, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | | | - Hoda Najjar
- Biomedical Sciences Department, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Layla Kamareddine
- Biomedical Sciences Department, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
17
|
Johansen MD, Herrmann JL, Kremer L. Non-tuberculous mycobacteria and the rise of Mycobacterium abscessus. Nat Rev Microbiol 2020; 18:392-407. [PMID: 32086501 DOI: 10.1038/s41579-020-0331-1] [Citation(s) in RCA: 495] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2020] [Indexed: 12/17/2022]
Abstract
Infections caused by non-tuberculous mycobacteria (NTM) are increasing globally and are notoriously difficult to treat due to intrinsic resistance of these bacteria to many common antibiotics. NTM are diverse and ubiquitous in the environment, with only a few species causing serious and often opportunistic infections in humans, including Mycobacterium abscessus. This rapidly growing mycobacterium is one of the most commonly identified NTM species responsible for severe respiratory, skin and mucosal infections in humans. It is often regarded as one of the most antibiotic-resistant mycobacteria, leaving us with few therapeutic options. In this Review, we cover the proposed infection process of M. abscessus, its virulence factors and host interactions and highlight the commonalities and differences of M. abscessus with other NTM species. Finally, we discuss drug resistance mechanisms and future therapeutic options. Taken together, this knowledge is essential to further our understanding of this overlooked and neglected global threat.
Collapse
Affiliation(s)
- Matt D Johansen
- Institut de Recherche en Infectiologie de Montpellier, Centre National de la Recherche Scientifique UMR 9004, Université de Montpellier, Montpellier, France
| | - Jean-Louis Herrmann
- Université Paris-Saclay, UVSQ, Inserm, Infection et Inflammation, Montigny-Le-Bretonneux, France.,AP-HP. GHU Paris Saclay, Hôpital Raymond Poincaré, Garches, France
| | - Laurent Kremer
- Institut de Recherche en Infectiologie de Montpellier, Centre National de la Recherche Scientifique UMR 9004, Université de Montpellier, Montpellier, France. .,Inserm, Institut de Recherche en Infectiologie de Montpellier, Montpellier, France.
| |
Collapse
|
18
|
Horizontal Gene Transfer of Short-Chain Dehydrogenase Coding Genes Contribute to the Biofilm Formation and Pathogenicity on Mycobacterium grossiae sp. nov. PB739 T (=DSM 104744 T). Curr Microbiol 2020; 77:528-533. [PMID: 31907602 DOI: 10.1007/s00284-019-01832-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/29/2019] [Indexed: 10/25/2022]
Abstract
Mycobacterium grossiae sp. nov. of type strain PB739T is a Gram-positive acid-alcohol-fast rod-shaped bacterium, which was recently isolated from a 76-year-old male who suffered from a 1-year history of hemoptysis. This strain was described as novel species in Mycobacterium genus. In this study, its genome was completely sequenced by PacBio technology, analyzed, and compared with other selected complete genome sequences of Mycobacterium to elucidate the distinct pathogenic features of the strain. The genomic analysis revealed that the genome of PB739T consists of one circular DNA chromosome of 5,637,923 bp with a GC content of 70.48% and one plasmid of 43,679 bp with a GC content of 66.24%. The entire genome contains 5434 predicted coding genes, 48 tRNAs, and 6 rRNA genes. Genome and comparative genomics against M. grossiae SCH identified three tandem short-chain dehydrogenase (SDR) genes which only exist in PB739T. These three tandem SDR genes locate in a Genomic island which was identified by Island Viewer. These SDR genes were predicted to be horizontally transferred from a Streptomyces ancestor based on phylogeny. Analysis of the mutant ΔSDR confirmed the relationship between these tandem genes with biofilm and pathogenicity. This report will provide us with an extended understanding of M. grossiae at the genomic level and would be helpful for understanding the evolution of Mycobacterium genus.
Collapse
|
19
|
Synergistic Efficacy of β-Lactam Combinations against Mycobacterium abscessus Pulmonary Infection in Mice. Antimicrob Agents Chemother 2019; 63:AAC.00614-19. [PMID: 31109979 DOI: 10.1128/aac.00614-19] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/11/2019] [Indexed: 01/31/2023] Open
Abstract
Mycobacterium abscessus is an emerging pathogen capable of causing invasive pulmonary infections in patients with chronic lung diseases. These infections are difficult to treat, necessitating prolonged multidrug therapy, which is further complicated by extensive intrinsic and acquired resistance exhibited by clinical M. abscessus isolates. Therefore, development of novel treatment regimens effective against drug-resistant strains is crucial. Prior studies have demonstrated synergistic efficacy of several β-lactams against M. abscessus in vitro; however, these combinations have never been tested in an animal model of M. abscessus pulmonary disease. We utilized a recently developed murine system of sustained M. abscessus lung infection delivered via an aerosol route to test the bactericidal efficacy of four novel dual β-lactam combinations and one β-lactam/β-lactamase inhibitor combination. All five of the novel combinations exhibited synergy and resulted in at least 6-log10 reductions in bacterial burden in the lungs of mice at 4 weeks compared to untreated controls (P = 0.038).
Collapse
|
20
|
Li Y, Spiropoulos J, Cooley W, Khara JS, Gladstone CA, Asai M, Bossé JT, Robertson BD, Newton SM, Langford PR. Galleria mellonella - a novel infection model for the Mycobacterium tuberculosis complex. Virulence 2018; 9:1126-1137. [PMID: 30067135 PMCID: PMC6086298 DOI: 10.1080/21505594.2018.1491255] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Animal models have long been used in tuberculosis research to understand disease pathogenesis and to evaluate novel vaccine candidates and anti-mycobacterial drugs. However, all have limitations and there is no single animal model which mimics all the aspects of mycobacterial pathogenesis seen in humans. Importantly mice, the most commonly used model, do not normally form granulomas, the hallmark of tuberculosis infection. Thus there is an urgent need for the development of new alternative in vivo models. The insect larvae, Galleria mellonella has been increasingly used as a successful, simple, widely available and cost-effective model to study microbial infections. Here we report for the first time that G. mellonella can be used as an infection model for members of the Mycobacterium tuberculosis complex. We demonstrate a dose-response for G. mellonella survival infected with different inocula of bioluminescent Mycobacterium bovis BCG lux, and demonstrate suppression of mycobacterial luminesence over 14 days. Histopathology staining and transmission electron microscopy of infected G. mellonella phagocytic haemocytes show internalization and aggregation of M. bovis BCG lux in granuloma-like structures, and increasing accumulation of lipid bodies within M. bovis BCG lux over time, characteristic of latent tuberculosis infection. Our results demonstrate that G. mellonella can act as a surrogate host to study the pathogenesis of mycobacterial infection and shed light on host-mycobacteria interactions, including latent tuberculosis infection.
Collapse
Affiliation(s)
- Yanwen Li
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK
| | - John Spiropoulos
- b Department of Pathology , Animal and Plant Health Agency , Addlestone , UK
| | - William Cooley
- b Department of Pathology , Animal and Plant Health Agency , Addlestone , UK
| | - Jasmeet Singh Khara
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK.,c Department of Pharmacy , National University of Singapore , Singapore
| | - Camilla A Gladstone
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK
| | - Masanori Asai
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK
| | - Janine T Bossé
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK
| | - Brian D Robertson
- d MRC Centre for Molecular Bacteriology and Infection, Department of Medicine , Imperial College London , London , UK
| | - Sandra M Newton
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK
| | - Paul R Langford
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK
| |
Collapse
|
21
|
Establishment and Validation of Galleria mellonella as a Novel Model Organism To Study Mycobacterium abscessus Infection, Pathogenesis, and Treatment. Antimicrob Agents Chemother 2018; 62:AAC.02539-17. [PMID: 29437630 DOI: 10.1128/aac.02539-17] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/01/2018] [Indexed: 11/20/2022] Open
Abstract
Treatment of Mycobacterium abscessus infections is extremely challenging due to its intrinsic resistance to most antibiotics, and research of pathogenesis is limited due to a lack of a practical in vivo model of infection. The objective of this study was to establish a simple in vivo model for M. abscessus infection, virulence, and drug testing in Galleria mellonella larvae. We inoculated larvae with M. abscessus bacteria and assessed histopathology, CFU count, and mortality with and without antibiotic treatment. We also constructed a luminescent, recombinant M. abscessus mutant, mDB158, and imaged infected larvae using the IVIS in vivo imaging system. M. abscessus proliferated and induced granuloma-like responses in infected larvae, leading to larval mortality. The G. mellonella model was further validated successfully by demonstration of the expected favorable antimicrobial effect of treatment with meropenem and the superiority of combination treatment (meropenem and tigecycline) over that with single agents. We then used IVIS imaging of larvae infected with luminescent M. abscessus, allowing live real-time assessment of bacterial load. We used this method to compare the antimicrobial effects of various antibiotics (meropenem, amikacin, linezolid, levofloxacin, etc.) on bacterial proliferation and larval survival. Meropenem and amikacin had the most favorable effects, correlating well with common clinical practice guidelines. These findings suggest G. mellonella to be an excellent in vivo model for research of M. abscessus infection, pathogenesis, and treatment. Luminescent M. abscessus and IVIS imaging further facilitates this model. Results obtained in this model clearly substantiated common clinical practice, thus validating the model as a predictor of treatment efficacy and outcome.
Collapse
|
22
|
Bernut A, Herrmann JL, Ordway D, Kremer L. The Diverse Cellular and Animal Models to Decipher the Physiopathological Traits of Mycobacterium abscessus Infection. Front Cell Infect Microbiol 2017; 7:100. [PMID: 28421165 PMCID: PMC5378707 DOI: 10.3389/fcimb.2017.00100] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/14/2017] [Indexed: 01/09/2023] Open
Abstract
Mycobacterium abscessus represents an important respiratory pathogen among the rapidly-growing non-tuberculous mycobacteria. Infections caused by M. abscessus are increasingly found in cystic fibrosis (CF) patients and are often refractory to antibiotic therapy. The underlying immunopathological mechanisms of pathogenesis remain largely unknown. A major reason for the poor advances in M. abscessus research has been a lack of adequate models to study the acute and chronic stages of the disease leading to delayed progress of evaluation of therapeutic efficacy of potentially active antibiotics. However, the recent development of cellular models led to new insights in the interplay between M. abscessus with host macrophages as well as with amoebae, proposed to represent the environmental host and reservoir for non-tuberculous mycobacteria. The zebrafish embryo has also appeared as a useful alternative to more traditional models as it recapitulates the vertebrate immune system and, due to its optical transparency, allows a spatio-temporal visualization of the infection process in a living animal. More sophisticated immunocompromised mice have also been exploited recently to dissect the immune and inflammatory responses to M. abscessus. Herein, we will discuss the limitations, advantages and potential offered by these various models to study the pathophysiology of M. abscessus infection and to assess the preclinical efficacy of compounds active against this emerging human pathogen.
Collapse
Affiliation(s)
- Audrey Bernut
- IRIM (ex-CPBS)-UMR 9004, Centre National de la Recherche Scientifique (CNRS), Infectious Disease Research Institute of Montpellier, Université de MontpellierMontpellier, France
| | - Jean-Louis Herrmann
- UMR 1173, Institut National de la Santé et de la Recherche Médicale, Université de Versailles Saint-Quentin-en-YvelinesMontigny-le-Bretonneux, France
| | - Diane Ordway
- Mycobacteria Research Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State UniversityFort Collins, CO, USA
| | - Laurent Kremer
- IRIM (ex-CPBS)-UMR 9004, Centre National de la Recherche Scientifique (CNRS), Infectious Disease Research Institute of Montpellier, Université de MontpellierMontpellier, France.,Institut National de la Santé et de la Recherche Médicale, IRIMMontpellier, France
| |
Collapse
|
23
|
Amikacin Pharmacokinetics/Pharmacodynamics in a Novel Hollow-Fiber Mycobacterium abscessus Disease Model. Antimicrob Agents Chemother 2015; 60:1242-8. [PMID: 26643339 DOI: 10.1128/aac.02282-15] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/29/2015] [Indexed: 11/20/2022] Open
Abstract
The treatment of pulmonary Mycobacterium abscessus disease is associated with very high failure rates and easily acquired drug resistance. Amikacin is the key drug in treatment regimens, but the optimal doses are unknown. No good preclinical model exists to perform formal pharmacokinetics/pharmacodynamics experiments to determine these optimal doses. We developed a hollow-fiber system model of M. abscessus disease and studied amikacin exposure effects and dose scheduling. We mimicked amikacin human pulmonary pharmacokinetics. Both amikacin microbial kill and acquired drug resistance were linked to the peak concentration-to-MIC ratios; the peak/MIC ratio associated with 80% of maximal kill (EC80) was 3.20. However, on the day of the most extensive microbial kill, the bacillary burden did not fall below the starting inoculum. We performed Monte Carlo simulations of 10,000 patients with pulmonary M. abscessus infection and examined the probability that patients treated with one of 6 doses from 750 mg to 4,000 mg would achieve or exceed the EC80. We also examined these doses for the ability to achieve a cumulative area under the concentration-time curve of 82,232 mg · h/liter × days, which is associated with ototoxicity. The standard amikacin doses of 750 to 1,500 mg a day achieved the EC80 in ≤ 21% of the patients, while a dose of 4 g/day achieved this in 70% of the patients but at the cost of high rates of ototoxicity within a month or two. The susceptibility breakpoint was an MIC of 8 to 16 mg/liter. Thus, amikacin, as currently dosed, has limited efficacy against M. abscessus. It is urgent that different antibiotics be tested using our preclinical model and new regimens developed.
Collapse
|
24
|
Bernut A, Dupont C, Sahuquet A, Herrmann JL, Lutfalla G, Kremer L. Deciphering and Imaging Pathogenesis and Cording of Mycobacterium abscessus in Zebrafish Embryos. J Vis Exp 2015. [PMID: 26382225 DOI: 10.3791/53130] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Zebrafish (Danio rerio) embryos are increasingly used as an infection model to study the function of the vertebrate innate immune system in host-pathogen interactions. The ease of obtaining large numbers of embryos, their accessibility due to external development, their optical transparency as well as the availability of a wide panoply of genetic/immunological tools and transgenic reporter line collections, contribute to the versatility of this model. In this respect, the present manuscript describes the use of zebrafish as an in vivo model system to investigate the chronology of Mycobacterium abscessus infection. This human pathogen can exist either as smooth (S) or rough (R) variants, depending on cell wall composition, and their respective virulence can be imaged and compared in zebrafish embryos and larvae. Micro-injection of either S or R fluorescent variants directly in the blood circulation via the caudal vein, leads to chronic or acute/lethal infections, respectively. This biological system allows high resolution visualization and analysis of the role of mycobacterial cording in promoting abscess formation. In addition, the use of fluorescent bacteria along with transgenic zebrafish lines harbouring fluorescent macrophages produces a unique opportunity for multi-color imaging of the host-pathogen interactions. This article describes detailed protocols for the preparation of homogenous M. abscessus inoculum and for intravenous injection of zebrafish embryos for subsequent fluorescence imaging of the interaction with macrophages. These techniques open the avenue to future investigations involving mutants defective in cord formation and are dedicated to understand how this impacts on M. abscessus pathogenicity in a whole vertebrate.
Collapse
Affiliation(s)
- Audrey Bernut
- Dynamique des Interactions Membranaires Normales et Pathologiques, CNRS, UMR 535, Université Montpellier; Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé, CNRS, FRE 3689, Université Montpellier
| | - Christian Dupont
- Dynamique des Interactions Membranaires Normales et Pathologiques, CNRS, UMR 535, Université Montpellier; Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé, CNRS, FRE 3689, Université Montpellier
| | - Alain Sahuquet
- Dynamique des Interactions Membranaires Normales et Pathologiques, CNRS, UMR 535, Université Montpellier
| | - Jean-Louis Herrmann
- Unité de Formation et de Recherche des Sciences de la Santé, EA3647-EPIM, Université Versailles St Quentin
| | - Georges Lutfalla
- Dynamique des Interactions Membranaires Normales et Pathologiques, CNRS, UMR 535, Université Montpellier;
| | - Laurent Kremer
- Dynamique des Interactions Membranaires Normales et Pathologiques, CNRS, UMR 535, Université Montpellier; Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé, CNRS, FRE 3689, Université Montpellier;
| |
Collapse
|
25
|
Howard ST. Recent progress towards understanding genetic variation in the Mycobacterium abscessus complex. Tuberculosis (Edinb) 2014; 93 Suppl:S15-20. [PMID: 24388643 DOI: 10.1016/s1472-9792(13)70005-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mycobacterium abscessus is an emerging cause of respiratory disease and soft tissue infections. Whole genome sequencing and other molecular approaches are enhancing our understanding of outbreaks, antibiotic resistance mechanisms, and virulence properties, and of the phylogeny of the M. abscessus complex. Infection models are providing further insights into factors such as colony phenotype that impact host-pathogen interactions. This paper reviews recent developments in our understanding of genetic variation in M. abscessus and the potential relevance for disease and treatment.
Collapse
Affiliation(s)
- Susan T Howard
- Department of Microbiology, University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA.
| |
Collapse
|
26
|
In vivo assessment of drug efficacy against Mycobacterium abscessus using the embryonic zebrafish test system. Antimicrob Agents Chemother 2014; 58:4054-63. [PMID: 24798271 DOI: 10.1128/aac.00142-14] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mycobacterium abscessus is responsible for a wide spectrum of clinical syndromes and is one of the most intrinsically drug-resistant mycobacterial species. Recent evaluation of the in vivo therapeutic efficacy of the few potentially active antibiotics against M. abscessus was essentially performed using immunocompromised mice. Herein, we assessed the feasibility and sensitivity of fluorescence imaging for monitoring the in vivo activity of drugs against acute M. abscessus infection using zebrafish embryos. A protocol was developed where clarithromycin and imipenem were directly added to water containing fluorescent M. abscessus-infected embryos in a 96-well plate format. The status of the infection with increasing drug concentrations was visualized on a spatiotemporal level. Drug efficacy was assessed quantitatively by measuring the index of protection, the bacterial burden (CFU), and the number of abscesses through fluorescence measurements. Both drugs were active in infected embryos and were capable of significantly increasing embryo survival in a dose-dependent manner. Protection from bacterial killing correlated with restricted mycobacterial growth in the drug-treated larvae and with reduced pathophysiological symptoms, such as the number of abscesses within the brain. In conclusion, we present here a new and efficient method for testing and compare the in vivo activity of two clinically relevant drugs based on a fluorescent reporter strain in zebrafish embryos. This approach could be used for rapid determination of the in vivo drug susceptibility profile of clinical isolates and to assess the preclinical efficacy of new compounds against M. abscessus.
Collapse
|
27
|
Oh CT, Moon C, Park OK, Kwon SH, Jang J. Novel drug combination for Mycobacterium abscessus disease therapy identified in a Drosophila infection model. J Antimicrob Chemother 2014; 69:1599-607. [PMID: 24519481 DOI: 10.1093/jac/dku024] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Mycobacterium abscessus is known to be the most drug-resistant Mycobacterium and accounts for ∼80% of pulmonary infections caused by rapidly growing mycobacteria. This study reports a new Drosophila melanogaster-M. abscessus infection model that can be used as an in vivo efficacy model for anti-M. abscessus drug potency assessment. METHODS D. melanogaster were challenged with M. abscessus, and infected flies were fed with a fly medium containing tigecycline, clarithromycin, linezolid, clofazimine, moxifloxacin, amikacin, cefoxitin, dinitrobenzamide or metronidazole at different concentrations (0, 100 and 500 mg/L). The survival rates of infected flies were plotted and bacterial colonization/dissemination in fly bodies was monitored by cfu determination and green fluorescent protein epifluorescence. RESULTS The D. melanogaster-M. abscessus model enabled an assessment of the effectiveness of antibiotic treatment. Tigecycline was the best drug for extending the lifespan of M. abscessus-infected Drosophila, followed by clarithromycin and linezolid. Several different combinations of tigecycline, linezolid and clarithromycin were tested to determine the best combination. Tigecycline (25 mg/L) plus linezolid (500 mg/L) was the best drug combination and its efficacy was superior to conventional regimens, not only in prolonging infected fly survival but also against M. abscessus colonization and dissemination. CONCLUSIONS This D. melanogaster-M. abscessus infection/curing methodology may be useful for the rapid evaluation of potential drug candidates. In addition, new combinations using tigecycline and linezolid should be considered as possible next-generation combination therapies to be assessed in higher organisms.
Collapse
Affiliation(s)
- Chun-Taek Oh
- Institute Pasteur Korea, Seongnam-si, Gyeonggi-do, Korea
| | - Cheol Moon
- Department of Clinical Laboratory Science, Semyung University, Jecheon, Chungbuk, Korea
| | - Ok Kyu Park
- Division of Analytical Bio-imaging, Chuncheon Center, Korea Basic Science Institute, Chuncheon, Gangwon-do, Korea
| | - Seung-Hae Kwon
- Division of Analytical Bio-imaging, Chuncheon Center, Korea Basic Science Institute, Chuncheon, Gangwon-do, Korea
| | - Jichan Jang
- Institute Pasteur Korea, Seongnam-si, Gyeonggi-do, Korea
| |
Collapse
|
28
|
Panayidou S, Ioannidou E, Apidianakis Y. Human pathogenic bacteria, fungi, and viruses in Drosophila: disease modeling, lessons, and shortcomings. Virulence 2014; 5:253-69. [PMID: 24398387 DOI: 10.4161/viru.27524] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Drosophila has been the invertebrate model organism of choice for the study of innate immune responses during the past few decades. Many Drosophila-microbe interaction studies have helped to define innate immunity pathways, and significant effort has been made lately to decipher mechanisms of microbial pathogenesis. Here we catalog 68 bacterial, fungal, and viral species studied in flies, 43 of which are relevant to human health. We discuss studies of human pathogens in flies revealing not only the elicitation and avoidance of immune response but also mechanisms of tolerance, host tissue homeostasis, regeneration, and predisposition to cancer. Prominent among those is the emerging pattern of intestinal regeneration as a defense response induced by pathogenic and innocuous bacteria. Immunopathology mechanisms and many microbial virulence factors have been elucidated, but their relevance to human health conventionally necessitates validation in mammalian models of infection.
Collapse
Affiliation(s)
- Stavria Panayidou
- Department of Biological Sciences; University of Cyprus; Nicosia, Cyprus
| | - Eleni Ioannidou
- Department of Biological Sciences; University of Cyprus; Nicosia, Cyprus
| | | |
Collapse
|