1
|
Feizi S, Awad M, Ramezanpour M, Cooksley C, Murphy W, Prestidge CA, Psaltis AJ, Wormald PJ, Barry S, Vreugde S. Promoting the Efficacy of Deferiprone-Gallium-Protoporphyrin (IX) against Mycobacterium abscessus Intracellular Infection with Lipid Liquid Crystalline Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2024; 16:70274-70283. [PMID: 39660476 DOI: 10.1021/acsami.4c15843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Nontuberculous mycobacteria (NTM) are among the recalcitrant bacterial strains that cause difficult-to-treat infections for patients with chronic underlying pulmonary conditions. The bacteria's intrinsic resistance to various antibiotics and their ability to infect macrophages enable them to overcome both the host immune response and standard antibiotics. Unconventional approaches to treating NTM-mediated infections are required. Using the heme mimic agent gallium protoporphyrin (GaPP) and the iron chelator deferiprone (DEF) in combination has been proven as an effective strategy against different bacteria including NTM in vitro. To enable more effective delivery and promote the activity of DEF/GaPP against intracellular NTM infections, both compounds are loaded in lipid liquid crystalline nanoparticles (LCNP). GaPP and DEF are sufficiently entrapped in LCNP with entrapment efficiency of 98% ± 2.1 and 39.4% ± 4.2, respectively. DEF/GaPP LCNP has an average diameter of 171 nm ± 10.2 with a uniform size distribution. DEF/GaPP LCNP reduces the viability of Mycobacterium abscessus intracellular infection by 3.34 log10 in comparison to the control group and is significantly more efficacious than nonformulated DEF/GaPP. Furthermore, DEF/GaPP LCNP is nontoxic to human bronchial epithelial cells in vitro. These findings are envisaged to pave the way for future progress in eradicating NTM-mediated infections.
Collapse
Affiliation(s)
- Sholeh Feizi
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Adelaide 5011, Australia
- The Department of Surgery, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia
| | - Muhammed Awad
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Adelaide 5011, Australia
- The Department of Surgery, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia
| | - Mahnaz Ramezanpour
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Adelaide 5011, Australia
- The Department of Surgery, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia
| | - Clare Cooksley
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Adelaide 5011, Australia
- The Department of Surgery, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia
| | - William Murphy
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Adelaide 5011, Australia
- The Department of Surgery, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia
| | - Clive A Prestidge
- Centre for Pharmaceutical Innovation, University of South Australia, Clinical and Health Sciences, Adelaide 5000, Australia
| | - Alkis J Psaltis
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Adelaide 5011, Australia
- The Department of Surgery, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia
| | - Peter-John Wormald
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Adelaide 5011, Australia
- The Department of Surgery, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia
| | - Simone Barry
- Precision Medicine Theme, South Australian Health and Medical Institute, Adelaide 5000, South Australia, Australia
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide 5000, South Australia, Australia
| | - Sarah Vreugde
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Adelaide 5011, Australia
- The Department of Surgery, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia
| |
Collapse
|
2
|
Naruoka S, Sakata S, Kawabata S, Hashiguchi Y, Daikoku E, Sakaguchi S, Okazaki F, Yoshikawa K, Rawls JF, Nakano T, Hirose Y, Ono F. A zebrafish gene with sequence similarities to human uromodulin and GP2 displays extensive evolutionary diversification among teleost and confers resistance to bacterial infection. Heliyon 2024; 10:e37510. [PMID: 39309883 PMCID: PMC11415648 DOI: 10.1016/j.heliyon.2024.e37510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
In the process of investigating synaptic changes happening to mutants lacking postsynaptic receptors in the neuromuscular junction, we focused on a hitherto uncharacterized zebrafish gene zgc153932 whose expression was increased in the RNAseq and droplet digital PCR (ddPCR) analysis of a paralyzed mutant sofa potato. The zgc153932 gene which we named omcin5 (omc5) showed amino acid sequence similarity to human uromodulin and GP2, which are expressed in epithelial cells of the kidney and the gut respectively and bind to bacteria pili. omc5 had 14 paralogues in a ∼400 KB region on the chromosome 12 of the zebrafish genome. These genes, named omcin1 through 15, constitute a gene cluster which presumably arose from recent gene duplications in the zebrafish lineage. An antibody raised against the epitope common to 6-9 genes in the omcin family revealed expression in the cloaca of 1 day post fertilization (dpf) embryos which broadened to the urinary and digestive tracts by 5 dpf. Expression of omc5 was increased by exposure of embryos to Escherichia coli (E. coli). Survival of omc5 mutant embryos was shortened in the presence of E. coli, or when they were not maintained in germ-free conditions. Adults omc5 mutants also exhibited susceptibility to infection. Other teleost species which had omcin-like genes in their genomes showed a range of gene duplication, resulting in clusters of 1 to >15 omcin-like genes. We hereby identified a new gene family specific to teleost that include a microbial induced gene which confers resistance to bacterial infection.
Collapse
Affiliation(s)
- Shiori Naruoka
- Department of Physiology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Japan
| | - Souhei Sakata
- Department of Physiology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Japan
| | - Shigeru Kawabata
- Department of Pathology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Japan
| | - Yasuyuki Hashiguchi
- Department of Biology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Japan
| | - Eriko Daikoku
- Department of Physiology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Japan
| | - Shoichi Sakaguchi
- Department of Microbiology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Japan
| | - Fumiyoshi Okazaki
- Department of Life Sciences, Graduate School of Bioresources, Mie University, Japan
| | - Kento Yoshikawa
- Department of Physiology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Japan
| | - John F. Rawls
- Department of Molecular Genetics & Microbiology, Duke Microbiome Center, Duke University School of Medicine, USA
| | - Takashi Nakano
- Department of Microbiology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Japan
| | - Yoshinobu Hirose
- Department of Pathology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Japan
| | - Fumihito Ono
- Department of Physiology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Japan
| |
Collapse
|
3
|
Johansen MD, Spaink HP, Oehlers SH, Kremer L. Modeling nontuberculous mycobacterial infections in zebrafish. Trends Microbiol 2024; 32:663-677. [PMID: 38135617 DOI: 10.1016/j.tim.2023.11.011] [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: 10/24/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023]
Abstract
The incidence of infections due to nontuberculous mycobacteria (NTM) has increased rapidly in recent years, surpassing tuberculosis in developed countries. Due to inherent antimicrobial resistance, NTM infections are particularly difficult to treat with low cure rates. There is an urgent need to understand NTM pathogenesis and to develop novel therapeutic approaches for the treatment of NTM diseases. Zebrafish have emerged as an excellent animal model due to genetic amenability and optical transparency during embryonic development, allowing spatiotemporal visualization of host-pathogen interactions. Furthermore, adult zebrafish possess fully functional innate and adaptive immunity and recapitulate important pathophysiological hallmarks of mycobacterial infection. Here, we report recent breakthroughs in understanding the hallmarks of NTM infections using the zebrafish model.
Collapse
Affiliation(s)
- Matt D Johansen
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia
| | - Herman P Spaink
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Stefan H Oehlers
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Laurent Kremer
- Centre National de la Recherche Scientifique, UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France; INSERM, IRIM, 34293 Montpellier, France.
| |
Collapse
|
4
|
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
|
5
|
Shekhar, Alcaraz M, Anand A, Sharma RK, Kremer L, Kumar V. Cu-promoted synthesis of triclosan-Mannich and Glaser adducts: anti-mycobacterial evaluation with in silico validations. Future Med Chem 2024; 16:949-961. [PMID: 38910577 DOI: 10.4155/fmc-2023-0298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/26/2024] [Indexed: 06/25/2024] Open
Abstract
Aim: The WHO, Global tuberculosis report 2022 estimated number of tuberculosis (TB) cases reached 10.6 million in 2021, reflecting a 4.5% increase compared with the 10.1 million reported in 2020. The incidence rate of TB showed 3.6% rise from 2020 to 2021. Results/methodology: This manuscript discloses Cu-promoted single pot A3-coupling between triclosan (TCS)-based alkyne, formaldehyde and secondary amines to yield TCS-based Mannich adducts. Additionally, the coupling of TCS-alkynes in the presence of Cu(OAc)2 afforded the corresponding homodimers. Among tested compounds, the most potent one in the series 11 exhibited fourfold higher potency than rifabutin against drug-resistant Mycobacterium abscessus. The selectivity index was also substantially improved, being 26 (day 1) and 15 (day 3), which is four-times better than TCS.
Collapse
Affiliation(s)
- Shekhar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, Punjab, 143005, India
| | - Matthéo Alcaraz
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, Montpellier, 34293, France
| | - Amit Anand
- Department of Chemistry, Khalsa college, Amritsar, Punjab, 143005, India
| | - Rajni Kant Sharma
- Department of Chemistry, College of Basic Science & Humanities CCS, Haryana Agricultural University, Hisar, Haryana, India
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, Montpellier, 34293, France
- INSERM, IRIM, Montpellier, 34293, France
| | - Vipan Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, Punjab, 143005, India
| |
Collapse
|
6
|
Habjan E, Schouten GK, Speer A, van Ulsen P, Bitter W. Diving into drug-screening: zebrafish embryos as an in vivo platform for antimicrobial drug discovery and assessment. FEMS Microbiol Rev 2024; 48:fuae011. [PMID: 38684467 PMCID: PMC11078164 DOI: 10.1093/femsre/fuae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/24/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024] Open
Abstract
The rise of multidrug-resistant bacteria underlines the need for innovative treatments, yet the introduction of new drugs has stagnated despite numerous antimicrobial discoveries. A major hurdle is a poor correlation between promising in vitro data and in vivo efficacy in animal models, which is essential for clinical development. Early in vivo testing is hindered by the expense and complexity of existing animal models. Therefore, there is a pressing need for cost-effective, rapid preclinical models with high translational value. To overcome these challenges, zebrafish embryos have emerged as an attractive model for infectious disease studies, offering advantages such as ethical alignment, rapid development, ease of maintenance, and genetic manipulability. The zebrafish embryo infection model, involving microinjection or immersion of pathogens and potential antibiotic hit compounds, provides a promising solution for early-stage drug screening. It offers a cost-effective and rapid means of assessing the efficacy, toxicity and mechanism of action of compounds in a whole-organism context. This review discusses the experimental design of this model, but also its benefits and challenges. Additionally, it highlights recently identified compounds in the zebrafish embryo infection model and discusses the relevance of the model in predicting the compound's clinical potential.
Collapse
Affiliation(s)
- Eva Habjan
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Gina K Schouten
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Alexander Speer
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Peter van Ulsen
- Section Molecular Microbiology of A-LIFE, Vrije Universiteit, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Section Molecular Microbiology of A-LIFE, Vrije Universiteit, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
7
|
Nguyen TQ, Heo BE, Jeon S, Ash A, Lee H, Moon C, Jang J. Exploring antibiotic resistance mechanisms in Mycobacterium abscessus for enhanced therapeutic approaches. Front Microbiol 2024; 15:1331508. [PMID: 38380095 PMCID: PMC10877060 DOI: 10.3389/fmicb.2024.1331508] [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: 11/01/2023] [Accepted: 01/17/2024] [Indexed: 02/22/2024] Open
Abstract
Mycobacterium abscessus, a leading cause of severe lung infections in immunocompromised individuals, poses significant challenges for current therapeutic strategies due to resistance mechanisms. Therefore, understanding the intrinsic and acquired antibiotic resistance of M. abscessus is crucial for effective treatment. This review highlights the mechanisms employed by M. abscessus to sustain antibiotic resistance, encompassing not only conventional drugs but also newly discovered drug candidates. This comprehensive analysis aims to identify novel entities capable of overcoming the notorious resistance exhibited by M. abscessus, providing insights for the development of more effective therapeutic interventions.
Collapse
Affiliation(s)
- Thanh Quang Nguyen
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Bo Eun Heo
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Seunghyeon Jeon
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Anwesha Ash
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Heehyun Lee
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Cheol Moon
- Department of Clinical Laboratory Science, Semyung University, Jecheon, Republic of Korea
| | - Jichan Jang
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
8
|
Kassegne L, Veziris N, Fraisse P. [A pharmacologic approach to treatment of Mycobacterium abscessus pulmonary disease]. Rev Mal Respir 2024; 41:29-42. [PMID: 38016833 DOI: 10.1016/j.rmr.2023.10.010] [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: 02/11/2023] [Accepted: 10/22/2023] [Indexed: 11/30/2023]
Abstract
Mycobacterium abscessus is a fast-growing non-tuberculous mycobacteria complex causing pulmonary infections, comprising the subspecies abscessus, massiliense and bolletii. Differences are based predominantly on natural inducible macrolide resistance, active in most Mycobacterium abscessus spp abscessus species and in Mycobacterium abscessus spp bolletii but inactive in Mycobacterium abscessus spp massiliense. Therapy consists in long-term treatment, combining multiple antibiotics. Prognosis is poor, as only 40% of patients experience cure. Pharmacodynamic and pharmacokinetic data on M. abscessus have recently been published, showing that therapy ineffectiveness might be explained by intrinsic bacterial resistance (macrolides…) and by the unfavorable pharmacokinetics of the recommended antibiotics. Other molecules and inhaled antibiotics are promising.
Collapse
Affiliation(s)
- L Kassegne
- Service de pneumologie, pôle de pathologie thoracique, nouvel hôpital civil, Strasbourg, France; Groupe pour l'enseignement et la recherche en pneumo-infectiologie de la SPLF, 66, boulevard Saint-Michel, 75006 Paris, France.
| | - N Veziris
- Département de bactériologie, Inserm U1135, Centre d'immunologie et des maladies infectieuses (CIMI-Paris), Centre national de référence des mycobactéries et de la résistance des mycobactéries aux antituberculeux, Groupe hospitalier AP-HP, Sorbonne université, site Saint-Antoine, Paris, France; Groupe pour l'enseignement et la recherche en pneumo-infectiologie de la SPLF, 66, boulevard Saint-Michel, 75006 Paris, France
| | - P Fraisse
- Service de pneumologie, pôle de pathologie thoracique, nouvel hôpital civil, Strasbourg, France; Groupe pour l'enseignement et la recherche en pneumo-infectiologie de la SPLF, 66, boulevard Saint-Michel, 75006 Paris, France
| |
Collapse
|
9
|
Carneiro S, Pinto M, Silva S, Santos A, Rodrigues I, Santos D, Duarte S, Vieira L, Gomes JP, Macedo R. Genome-Scale Characterization of Mycobacterium abscessus Complex Isolates from Portugal. Int J Mol Sci 2023; 24:15402. [PMID: 37895081 PMCID: PMC10606986 DOI: 10.3390/ijms242015402] [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: 09/27/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
The Mycobacterium abscessus complex (MABC) is an emerging, difficult to treat, multidrug-resistant nontuberculous mycobacteria responsible for a wide spectrum of infections and associated with an increasing number of cases worldwide. Dominant circulating clones (DCCs) of MABC have been genetically identified as groups of strains associated with higher prevalence, higher levels of antimicrobial resistance, and worse clinical outcomes. To date, little is known about the genomic characteristics of MABC species circulating in Portugal. Here, we examined the genetic diversity and antimicrobial resistance profiles of 30 MABC strains isolated between 2014 and 2022 in Portugal. The genetic diversity of circulating MABC strains was assessed through a gene-by-gene approach (wgMLST), allowing their subspecies differentiation and the classification of isolates into DCCs. Antimicrobial resistance profiles were defined using phenotypic, molecular, and genomic approaches. The majority of isolates were resistant to at least two antimicrobials, although a poor correlation between phenotype and genotype data was observed. Portuguese genomes were highly diverse, and data suggest the existence of MABC lineages with potential international circulation or cross-border transmission. This study highlights the genetic diversity and antimicrobial resistance profile of circulating MABC isolates in Portugal while representing the first step towards the implementation of a genomic-based surveillance system for MABC at the Portuguese NIH.
Collapse
Affiliation(s)
- Sofia Carneiro
- National Reference Laboratory for Mycobacteria, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), 1649-016 Lisbon, Portugal; (S.C.); (A.S.)
- Department of Life Science, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Lisbon, Portugal
| | - Miguel Pinto
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), 1649-016 Lisbon, Portugal; (M.P.); (J.P.G.)
| | - Sónia Silva
- National Reference Laboratory for Mycobacteria, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), 1649-016 Lisbon, Portugal; (S.C.); (A.S.)
| | - Andrea Santos
- National Reference Laboratory for Mycobacteria, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), 1649-016 Lisbon, Portugal; (S.C.); (A.S.)
| | - Irene Rodrigues
- National Reference Laboratory for Mycobacteria, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), 1649-016 Lisbon, Portugal; (S.C.); (A.S.)
| | - Daniela Santos
- Technology and Innovation Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), 1649-016 Lisbon, Portugal; (D.S.); (S.D.)
| | - Sílvia Duarte
- Technology and Innovation Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), 1649-016 Lisbon, Portugal; (D.S.); (S.D.)
| | - Luís Vieira
- Technology and Innovation Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), 1649-016 Lisbon, Portugal; (D.S.); (S.D.)
| | - João Paulo Gomes
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), 1649-016 Lisbon, Portugal; (M.P.); (J.P.G.)
- Veterinary and Animal Research Centre (CECAV), Faculty of Veterinary Medicine, Lusófona University, 376 Campo Grande, 1749-024 Lisbon, Portugal
| | - Rita Macedo
- National Reference Laboratory for Mycobacteria, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), 1649-016 Lisbon, Portugal; (S.C.); (A.S.)
| |
Collapse
|
10
|
Nie W, Gao S, Su L, Liu L, Geng R, You Y, Chu N. Antibacterial activity of the novel compound Sudapyridine (WX-081) against Mycobacterium abscessus. Front Cell Infect Microbiol 2023; 13:1217975. [PMID: 37662015 PMCID: PMC10471480 DOI: 10.3389/fcimb.2023.1217975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 07/18/2023] [Indexed: 09/05/2023] Open
Abstract
Objective This study aimed to investigate sudapyridine (WX-081) antibacterial activity against Mycobacterium abscessus in vitro and its effect on in vivo bacterial growth and host survival using a zebrafish model of M. abscessus infection. Methods WX-081 in vitro antibacterial activity was assessed based on growth inhibition of M. abscessus standard strain ATCC19977 and 36 clinical isolates. Maximum tolerated concentrations (MTCs) of WX-081, bedaquiline, and azithromycin and inhibition of M. abscessus growth were assessed in vivo after fluorescently labelled bacilli and drugs were injected into zebrafish. Bacterial counts were analysed using one-way ANOVA and fluorescence intensities of zebrafish tissues were analysed and expressed as the mean ± SE. Moreover, Kaplan-Meier survival analysis was conducted to assess intergroup differences in survival of M. abscessus-infected zebrafish treated with different drug concentrations using a log-rank test, with a p value <0.05 indicating a difference was statistically significant. Results Drug sensitivity testing of M. abscessus standard strain ATCC19977 and 36 clinical isolates revealed MICs ranging from 0.12-0.96 µg/mL and MIC50 and MIC90 values of 0.48 µg/mL and 0.96 µg/mL, respectively. Fluorescence intensities of M. abscessus-infected zebrafish tissues was lower after treatment with the WX-081 MTC (62.5 µg/mL) than after treatment with the azithromycin MTC (62.5 µg/mL) and the bedaquiline MTC (15.6 µg/mL). When the concentration of WX-081 increased from 1.95µg/mL to 1/8 MTC(7.81µg/mL), the survival rate of zebrafish at 4-9 dpf decreased from 90.00% to 81.67%. Conclusion WX-081 effectively inhibited M. abscessus growth in vitro and in vivo and prolonged survival of M. abscessus-infected zebrafish, thus indicating that WX-081 holds promise as a clinical treatment for M. abscessus infection.
Collapse
Affiliation(s)
- Wenjuan Nie
- Tuberculosis Department, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Tuberculosis Department, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Shan Gao
- Tuberculosis Department, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Tuberculosis Department, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Lei Su
- Tuberculosis Department, Henan Anyang City Tuberculosis Prevention and Control Institute, Anyang, China
| | - Lina Liu
- Tuberculosis Department, Hengshui Third People’s Hospital, Hengshui, China
| | - Ruixue Geng
- Tuberculosis Department, Hohhot Second Hospital, Hohhot, China
| | - Yingxia You
- Tuberculosis Department, Zhengzhou Sixth People’s Hospital, Zhengzhou, China
| | - Naihui Chu
- Tuberculosis Department, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Tuberculosis Department, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| |
Collapse
|
11
|
Gao S, Nie W, Liu L, Su L, You Y, Geng R, Chu N. Antibacterial activity of the novel oxazolidinone contezolid (MRX-I) against Mycobacterium abscessus. Front Cell Infect Microbiol 2023; 13:1225341. [PMID: 37655300 PMCID: PMC10465794 DOI: 10.3389/fcimb.2023.1225341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/18/2023] [Indexed: 09/02/2023] Open
Abstract
Objective To evaluate contezolid (MRX-I) antibacterial activity against Mycobacterium abscessus in vitro and in vivo and to assess whether MRX-I treatment can prolong survival of infected zebrafish. Methods MRX-I inhibitory activity against M. abscessus in vitro was assessed by injecting MRX-I into zebrafish infected with green fluorescent protein-labelled M. abscessus. Thereafter, infected zebrafish were treated with azithromycin (AZM), linezolid (LZD) or MRX-I then maximum tolerated concentrations (MTCs) of drugs were determined based on M. abscessus growth inhibition using one-way ANOVA. Linear trend analysis of CFU counts and fluorescence intensities (mean ± SE values) was performed to detect linear relationships between MRX-I, AZM and LZD concentrations and these parameters. Results MRX-I anti-M. abscessus minimum inhibitory concentration (MIC) and MTC were 16 μg/mL and 15.6 μg/mL, respectively. MRX-I MTC-treated zebrafish fluorescence intensities were significantly lower than respective LZD group intensities (whole-body: 439040 ± 3647 vs. 509184 ± 23064, p < 0.01); head: 74147 ± 2175 vs. 95996 ± 8054, p < 0.05). As MRX-I concentration was increased from 0.488 μg/mL to 15.6 μg/mL, zebrafish whole-body, head and heart fluorescence intensities decreased. Statistically insignificant differences between the MRX-I MTC group survival rate (78.33%) vs. corresponding rates of the 62.5 μg/mL-treated AZM MTC group (88.33%, p > 0.05) and the 15.6 μg/mL-treated LZD MTC group (76.67%, p > 0.05) were observed. Conclusion MRX-I effectively inhibited M. abscessus growth and prolonged zebrafish survival when administered to M. abscessus-infected zebrafish, thus demonstrating that MRX-I holds promise as a clinical treatment for human M. abscessus infections.
Collapse
Affiliation(s)
- Shan. Gao
- Tuberculosis Department, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- Tuberculosis Department, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Wenjuan. Nie
- Tuberculosis Department, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- Tuberculosis Department, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Lina. Liu
- Tuberculosis Department, Hengshui Third People’s Hospital, Hengshui, China
| | - Lei. Su
- Tuberculosis Department, Henan Province Anyang City Tuberculosis Prevention and Control Institute, Anyang, China
| | - Yingxia. You
- Tuberculosis Department, Zhengzhou Sixth People’s Hospital, Zhengzhou, China
| | - Ruixue. Geng
- Tuberculosis Department, Hohhot Second Hospital, Hohhot, China
| | - Naihui Chu
- Tuberculosis Department, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- Tuberculosis Department, Beijing Chest Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Fries F, Kany AM, Rasheed S, Hirsch AKH, Müller R, Herrmann J. Impact of Drug Administration Routes on the In Vivo Efficacy of the Natural Product Sorangicin a Using a Staphylococcus aureus Infection Model in Zebrafish Embryos. Int J Mol Sci 2023; 24:12791. [PMID: 37628971 PMCID: PMC10454396 DOI: 10.3390/ijms241612791] [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: 06/28/2023] [Revised: 08/05/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Staphylococcus aureus causes a wide range of infections, and it is one of the leading pathogens responsible for deaths associated with antimicrobial resistance, the rapid spread of which among S. aureus urges the discovery of new antibiotics. The evaluation of in vivo efficacy of novel drug candidates is usually performed using animal models. Recently, zebrafish (Danio rerio) embryos have become increasingly attractive in early drug discovery. Herein, we established a zebrafish embryo model of S. aureus infection for evaluation of in vivo efficacy of novel potential antimicrobials. A local infection was induced by microinjecting mCherry-expressing S. aureus Newman followed by treatment with reference antibiotics via microinjection into different injection sites as well as via waterborne exposure to study the impact of the administration route on efficacy. We successfully used the developed model to evaluate the in vivo activity of the natural product sorangicin A, for which common mouse models were not successful due to fast degradation in plasma. In conclusion, we present a novel screening platform for assessing in vivo activity at the antibiotic discovery stage. Furthermore, this work provides consideration for the choice of an appropriate administration route based on the physicochemical properties of tested drugs.
Collapse
Affiliation(s)
- Franziska Fries
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123 Saarbrücken, Germany; (F.F.); (A.M.K.); (S.R.); (A.K.H.H.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Andreas M. Kany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123 Saarbrücken, Germany; (F.F.); (A.M.K.); (S.R.); (A.K.H.H.); (R.M.)
| | - Sari Rasheed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123 Saarbrücken, Germany; (F.F.); (A.M.K.); (S.R.); (A.K.H.H.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Anna K. H. Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123 Saarbrücken, Germany; (F.F.); (A.M.K.); (S.R.); (A.K.H.H.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123 Saarbrücken, Germany; (F.F.); (A.M.K.); (S.R.); (A.K.H.H.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123 Saarbrücken, Germany; (F.F.); (A.M.K.); (S.R.); (A.K.H.H.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| |
Collapse
|
13
|
Leon-Icaza SA, Bagayoko S, Vergé R, Iakobachvili N, Ferrand C, Aydogan T, Bernard C, Sanchez Dafun A, Murris-Espin M, Mazières J, Bordignon PJ, Mazères S, Bernes-Lasserre P, Ramé V, Lagarde JM, Marcoux J, Bousquet MP, Chalut C, Guilhot C, Clevers H, Peters PJ, Molle V, Lugo-Villarino G, Cam K, Berry L, Meunier E, Cougoule C. Druggable redox pathways against Mycobacterium abscessus in cystic fibrosis patient-derived airway organoids. PLoS Pathog 2023; 19:e1011559. [PMID: 37619220 PMCID: PMC10449475 DOI: 10.1371/journal.ppat.1011559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 07/13/2023] [Indexed: 08/26/2023] Open
Abstract
Mycobacterium abscessus (Mabs) drives life-shortening mortality in cystic fibrosis (CF) patients, primarily because of its resistance to chemotherapeutic agents. To date, our knowledge on the host and bacterial determinants driving Mabs pathology in CF patient lung remains rudimentary. Here, we used human airway organoids (AOs) microinjected with smooth (S) or rough (R-)Mabs to evaluate bacteria fitness, host responses to infection, and new treatment efficacy. We show that S Mabs formed biofilm, and R Mabs formed cord serpentines and displayed a higher virulence. While Mabs infection triggers enhanced oxidative stress, pharmacological activation of antioxidant pathways resulted in better control of Mabs growth and reduced virulence. Genetic and pharmacological inhibition of the CFTR is associated with better growth and higher virulence of S and R Mabs. Finally, pharmacological activation of antioxidant pathways inhibited Mabs growth, at least in part through the quinone oxidoreductase NQO1, and improved efficacy in combination with cefoxitin, a first line antibiotic. In conclusion, we have established AOs as a suitable human system to decipher mechanisms of CF-driven respiratory infection by Mabs and propose boosting of the NRF2-NQO1 axis as a potential host-directed strategy to improve Mabs infection control.
Collapse
Affiliation(s)
- Stephen Adonai Leon-Icaza
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Salimata Bagayoko
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Romain Vergé
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Nino Iakobachvili
- M4i Nanoscopy Division, Maastricht University, Maastricht, Netherlands
| | - Chloé Ferrand
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Talip Aydogan
- Laboratory of Pathogen Host Interactions (LPHI), Université Montpellier, CNRS, Montpellier, France
| | - Célia Bernard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Angelique Sanchez Dafun
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Marlène Murris-Espin
- Service de Pneumologie, Hôpital Larrey, CHU de Toulouse, Toulouse, France
- Centre de ressource et de compétence pour la mucoviscidose de l’adulte (CRCM adulte), CHU de Toulouse, Toulouse, France
| | - Julien Mazières
- Service de Pneumologie, Hôpital Larrey, CHU de Toulouse, Toulouse, France
| | - Pierre Jean Bordignon
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Serge Mazères
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | | | - Victoria Ramé
- Imactiv-3D SAS, 1 Place Pierre POTIER, Toulouse, France
| | | | - Julien Marcoux
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Marie-Pierre Bousquet
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Christian Chalut
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Christophe Guilhot
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, Netherlands
| | - Peter J. Peters
- M4i Nanoscopy Division, Maastricht University, Maastricht, Netherlands
| | - Virginie Molle
- Laboratory of Pathogen Host Interactions (LPHI), Université Montpellier, CNRS, Montpellier, France
| | - Geanncarlo Lugo-Villarino
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Kaymeuang Cam
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Laurence Berry
- Laboratory of Pathogen Host Interactions (LPHI), Université Montpellier, CNRS, Montpellier, France
| | - Etienne Meunier
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| |
Collapse
|
14
|
Alcaraz M, Edwards TE, Kremer L. New therapeutic strategies for Mycobacterium abscessus pulmonary diseases - untapping the mycolic acid pathway. Expert Rev Anti Infect Ther 2023; 21:813-829. [PMID: 37314394 PMCID: PMC10529309 DOI: 10.1080/14787210.2023.2224563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/08/2023] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Treatment options against Mycobacterium abscessus infections are very limited. New compounds are needed to cure M. abscessus pulmonary diseases. While the mycolic acid biosynthetic pathway has been largely exploited for the treatment of tuberculosis, this metabolic process has been overlooked in M. abscessus, although it offers many potential drug targets for the treatment of this opportunistic pathogen. AREAS COVERED Herein, the authors review the role of the MmpL3 membrane protein and the enoyl-ACP reductase InhA involved in the transport and synthesis of mycolic acids, respectively. They discuss their importance as two major vulnerable drug targets in M. abscessus and report the activity of MmpL3 and InhA inhibitors. In particular, they focus on NITD-916, a direct InhA inhibitor against M. abscessus, particularly warranted in the context of multidrug resistance. EXPERT OPINION There is an increasing body of evidence validating the mycolic acid pathway as an attractive drug target to be further exploited for M. abscessus lung disease treatments. The NITD-916 studies provide a proof-of-concept that direct inhibitors of InhA are efficient in vitro, in macrophages and in zebrafish. Future work is now required to improve the activity and pharmacological properties of these inhibitors and their evaluation in pre-clinical models.
Collapse
Affiliation(s)
- Matthéo Alcaraz
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293, Montpellier, France
| | - Thomas E. Edwards
- UCB BioSciences, Bainbridge Island, WA 98109 USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA 98109 USA
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293, Montpellier, France
- INSERM, IRIM, 34293 Montpellier, France
| |
Collapse
|
15
|
Goh BC, Larsson S, Dam LC, Ling YHS, Chua WLP, Abirami R, Singh S, Ong JLE, Teo JWP, Ho P, Ingham PW, Pethe K, Dedon PC. Rifaximin potentiates clarithromycin against Mycobacterium abscessus in vitro and in zebrafish. JAC Antimicrob Resist 2023; 5:dlad052. [PMID: 37168836 PMCID: PMC10164658 DOI: 10.1093/jacamr/dlad052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/13/2023] [Indexed: 05/13/2023] Open
Abstract
Background Mycobacterium abscessus is a non-tuberculous mycobacterium (NTM) that causes chronic pulmonary infections. Because of its extensive innate resistance to numerous antibiotics, treatment options are limited, often resulting in poor clinical outcomes. Current treatment regimens usually involve a combination of antibiotics, with clarithromycin being the cornerstone of NTM treatments. Objectives To identify drug candidates that exhibit synergistic activity with clarithromycin against M. abscessus. Methods We performed cell-based phenotypic screening of a compound library against M. abscessus induced to become resistant to clarithromycin. Furthermore, we evaluated the toxicity and efficacy of the top compound in a zebrafish embryo infection model. Results The screen revealed rifaximin as a clarithromycin potentiator. The combination of rifaximin and clarithromycin was synergistic and bactericidal in vitro and potent in the zebrafish model. Conclusions The data indicate that the rifaximin/clarithromycin combination is promising to effectively treat pulmonary NTM infections.
Collapse
Affiliation(s)
- Boon Chong Goh
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore
| | - Simon Larsson
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Linh Chi Dam
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore
| | - Yan Han Sharon Ling
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore
| | - Wei Lin Patrina Chua
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore
| | - R Abirami
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore
| | - Samsher Singh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jun Long Ernest Ong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jeanette W P Teo
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore
| | - Peiying Ho
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore
| | - Philip W Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A*Star), Singapore, Singapore
| | | | | |
Collapse
|
16
|
Roquet-Banères F, Alcaraz M, Hamela C, Abendroth J, Edwards TE, Kremer L. In Vitro and In Vivo Efficacy of NITD-916 against Mycobacterium fortuitum. Antimicrob Agents Chemother 2023; 67:e0160722. [PMID: 36920188 PMCID: PMC10112203 DOI: 10.1128/aac.01607-22] [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: 12/01/2022] [Accepted: 02/16/2023] [Indexed: 03/16/2023] Open
Abstract
Mycobacterium fortuitum represents one of the most clinically relevant rapid-growing mycobacterial species. Treatments are complex due to antibiotic resistance and to severe side effects of effective drugs, prolonged time of treatment, and co-infection with other pathogens. Herein, we explored the activity of NITD-916, a direct inhibitor of the enoyl-ACP reductase InhA of the type II fatty acid synthase in Mycobacterium tuberculosis. We found that this compound displayed very low MIC values against a panel of M. fortuitum clinical strains and exerted potent antimicrobial activity against M. fortuitum in macrophages. Remarkably, the compound was also highly efficacious in a zebrafish model of infection. Short duration treatments were sufficient to significantly protect the infected larvae from M. fortuitum-induced killing, which correlated with reduced bacterial burdens and abscesses. Biochemical analyses demonstrated an inhibition of de novo synthesis of mycolic acids. Resolving the crystal structure of the InhAMFO in complex with NAD and NITD-916 confirmed that NITD-916 is a direct inhibitor of InhAMFO. Importantly, single nucleotide polymorphism leading to a G96S substitution in InhAMFO conferred high resistance levels to NITD-916, thus resolving its target in M. fortuitum. Overall, these findings indicate that NITD-916 is highly active against M. fortuitum both in vitro and in vivo and should be considered in future preclinical evaluations for the treatment of M. fortuitum pulmonary diseases.
Collapse
Affiliation(s)
- Françoise Roquet-Banères
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | - Matthéo Alcaraz
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | - Claire Hamela
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | - Jan Abendroth
- UCB BioSciences, Bainbridge Island, Washington, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
| | - Thomas E. Edwards
- UCB BioSciences, Bainbridge Island, Washington, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
| | - 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
| |
Collapse
|
17
|
Nicola F, Cirillo DM, Lorè NI. Preclinical murine models to study lung infection with Mycobacterium abscessus complex. Tuberculosis (Edinb) 2023; 138:102301. [PMID: 36603391 DOI: 10.1016/j.tube.2022.102301] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
Mycobacterium abscessus is a non-tuberculous mycobacterium (NTM) able to cause invasive pulmonary infections, named NTM pulmonary disease. The therapeutic approaches are limited, and infections are difficult to treat due to antibiotic resistance conferred by an impermeable cell wall, drug efflux pumps, or drug-modifying enzymes. The development of new therapeutics, intended as antimicrobials or drug limiting immunopathology, is urgently necessary. In this context, the preclinical murine models of M. abscessus represent a useful tool to validate and translate in vitro-proofed concepts. These in vivo models are essential for developing new targets and drugs, ameliorating our knowledge in combinatorial regimens of current existing antibiotic treatments, and repurposing existing drugs for new therapeutic options against M. abscessus infection. Thus, this review aims at providing an overview of the current state of the art of preclinical murine models to study M. abscessus lung infection and its exploitation for new therapeutic approaches. This review discusses the murine models available focusing on the different bacterial challenges (aerosol, intranasal, intratracheal, and intravenous administrations), murine genetic background, and additional bacterial related factors. Then, we discuss the successful preclinical models for M. abscessus respiratory infection exploited to study the efficacy and safety of new antimicrobials or to determine the best dosage and route of administration of existing drugs. Finally, we present the current murine models exploited to develop new therapeutic approaches to modulate the host immune response and limit immunopathological damage during M. abscessus lung disease. In conclusion, our review article provides an overview of current and available murine models to characterize acute or chronic infections and to study the outcome of new therapeutic strategies against M. abscessus lung infection.
Collapse
Affiliation(s)
- Francesca Nicola
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela M Cirillo
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicola I Lorè
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
18
|
Jeon SM, Kim YJ, Nguyen TQ, Cui J, Thi Bich Hanh B, Silwal P, Kim JK, Kim JM, Oh DC, Jang J, Jo EK. Ohmyungsamycin Promotes M1-like Inflammatory Responses to Enhance Host Defense against Mycobacteroides abscessus Infections. Virulence 2022; 13:1966-1984. [PMID: 36271707 DOI: 10.1080/21505594.2022.2138009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Ohmyungsamycin A (OMS) is a newly identified cyclic peptide that exerts antimicrobial effects against Mycobacterium tuberculosis. However, its role in nontuberculous mycobacteria (NTMs) infections has not been clarified. Mycobacteroides abscessus (Mabc) is a rapidly growing NTM that has emerged as a human pathogen in both immunocompetent and immunosuppressed individuals. In this study, we demonstrated that OMS had significant antimicrobial effects against Mabc infection in both immunocompetent and immunodeficient mice, and in macrophages. OMS treatment amplified Mabc-induced expression of M1-related proinflammatory cytokines and inducible nitric oxide synthase, and significantly downregulated arginase-1 expression in murine macrophages. In addition, OMS augmented Mabc-mediated production of mitochondrial reactive oxygen species (mtROS), which promoted M1-like proinflammatory responses in Mabc-infected macrophages. OMS-induced production of mtROS and nitric oxide was critical for OMS-mediated antimicrobial responses during Mabc infections. Notably, the combination of OMS and rifabutin had a synergistic effect on the antimicrobial responses against Mabc infections in vitro, in murine macrophages, and in zebrafish models in vivo. Collectively, these data strongly suggest that OMS may be an effective M1-like adjunctive therapeutic against Mabc infections, either alone or in combination with antibiotics.
Collapse
Affiliation(s)
- Sang Min Jeon
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Young Jae Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Thanh Quang Nguyen
- Division of Applied Life Science (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Jinsheng Cui
- Department of Microbiology, Keimyung University School of Medicine, Daegu, South Korea
| | - Bui Thi Bich Hanh
- Division of Applied Life Science (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, South Korea
| | - Jin-Man Kim
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Pathology, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Dong-Chan Oh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Jichan Jang
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University,Jinju, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| |
Collapse
|
19
|
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
|
20
|
Kam JY, Wright K, Britton WJ, Oehlers SH. Treatment of infection-induced vascular pathologies is protective against persistent rough morphotype Mycobacterium abscessus infection in zebrafish. Microb Pathog 2022; 167:105590. [PMID: 35588967 DOI: 10.1016/j.micpath.2022.105590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/25/2022] [Accepted: 05/10/2022] [Indexed: 11/30/2022]
Abstract
Mycobacterium abscessus infections are of increasing global prevalence and are often difficult to treat due to complex antibiotic resistance profiles. While there are similarities between the pathogenesis of M. abscessus and tuberculous mycobacteria, including granuloma formation and stromal remodelling, there are distinct molecular differences at the host-pathogen interface. Here we have used a zebrafish-M. abscessus model and host-directed therapies that were previously identified in the zebrafish-M. marinum model to identify potential host-directed therapies against M. abscessus infection. We find efficacy of anti-angiogenic and vascular normalizing therapies against rough M. abscessus infection, but no effect of anti-platelet drugs.
Collapse
Affiliation(s)
- Julia Y Kam
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Kathryn Wright
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia; Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia; The University of Sydney, Sydney Institute for Infectious Diseases, Camperdown, NSW, 2050, Australia; A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.
| |
Collapse
|
21
|
Daher W, Leclercq LD, Johansen MD, Hamela C, Karam J, Trivelli X, Nigou J, Guérardel Y, Kremer L. Glycopeptidolipid glycosylation controls surface properties and pathogenicity in Mycobacterium abscessus. Cell Chem Biol 2022; 29:910-924.e7. [PMID: 35358417 DOI: 10.1016/j.chembiol.2022.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/17/2021] [Accepted: 03/10/2022] [Indexed: 12/18/2022]
Abstract
Mycobacterium abscessus is an emerging and difficult-to-manage mycobacterial species that exhibits smooth (S) or rough (R) morphotypes. Disruption of glycopeptidolipid (GPL) production results in transition from S to R and severe lung disease. A structure-activity relationship study was undertaken to decipher the role of GPL glycosylation in morphotype transition and pathogenesis. Deletion of gtf3 uncovered the prominent role of the extra rhamnose in enhancing mannose receptor-mediated internalization of M. abscessus by macrophages. In contrast, the absence of the 6-deoxy-talose and the first rhamnose in mutants lacking gtf1 and gtf2, respectively, affected M abscessus phagocytosis but also resulted in the S-to-R transition. Strikingly, gtf1 and gtf2 mutants displayed a strong propensity to form cords and abscesses in zebrafish, leading to robust and lethal infection. Together, these results underscore the importance and differential contribution of GPL monosaccharides in promoting virulence and infection outcomes.
Collapse
Affiliation(s)
- Wassim Daher
- CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France; INSERM, IRIM, 34293 Montpellier, France
| | - Louis-David Leclercq
- Université de Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000 Lille, France
| | - Matt D Johansen
- CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France; Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia
| | - Claire Hamela
- CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France
| | - Jona Karam
- CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France
| | - Xavier Trivelli
- Université de Lille, CNRS, INRAE, Centrale Lille, Université d'Artois, FR 2638 - IMEC - Institut Michel-Eugène Chevreul, 59000 Lille, France
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, Toulouse, France
| | - Yann Guérardel
- Université de Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000 Lille, France; Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan.
| | - Laurent Kremer
- CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France; INSERM, IRIM, 34293 Montpellier, France.
| |
Collapse
|
22
|
Ferrell KC, Johansen MD, Triccas JA, Counoupas C. Virulence Mechanisms of Mycobacterium abscessus: Current Knowledge and Implications for Vaccine Design. Front Microbiol 2022; 13:842017. [PMID: 35308378 PMCID: PMC8928063 DOI: 10.3389/fmicb.2022.842017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/08/2022] [Indexed: 12/22/2022] Open
Abstract
Mycobacterium abscessus is a member of the non-tuberculous mycobacteria (NTM) group, responsible for chronic infections in individuals with cystic fibrosis (CF) or those otherwise immunocompromised. While viewed traditionally as an opportunistic pathogen, increasing research into M. abscessus in recent years has highlighted its continued evolution into a true pathogen. This is demonstrated through an extensive collection of virulence factors (VFs) possessed by this organism which facilitate survival within the host, particularly in the harsh environment of the CF lung. These include VFs resembling those of other Mycobacteria, and non-mycobacterial VFs, both of which make a notable contribution in shaping M. abscessus interaction with the host. Mycobacterium abscessus continued acquisition of VFs is cause for concern and highlights the need for novel vaccination strategies to combat this pathogen. An effective M. abscessus vaccine must be suitably designed for target populations (i.e., individuals with CF) and incorporate current knowledge on immune correlates of protection against M. abscessus infection. Vaccination strategies must also build upon lessons learned from ongoing efforts to develop novel vaccines for other pathogens, particularly Mycobacterium tuberculosis (M. tb); decades of research into M. tb has provided insight into unconventional and innovative vaccine approaches that may be applied to M. abscessus. Continued research into M. abscessus pathogenesis will be critical for the future development of safe and effective vaccines and therapeutics to reduce global incidence of this emerging pathogen.
Collapse
Affiliation(s)
- Kia C. Ferrell
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program, Centenary Institute, Sydney, NSW, Australia
- *Correspondence: Kia C. Ferrell,
| | - Matt D. Johansen
- Centre for Inflammation, Centenary Institute, University of Technology, Sydney, NSW, Australia
- Faculty of Science, School of Life Sciences, University of Technology, Sydney, NSW, Australia
| | - James A. Triccas
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Sydney Institute for Infectious Diseases and the Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Claudio Counoupas
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program, Centenary Institute, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases and the Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
- Claudio Counoupas,
| |
Collapse
|
23
|
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: 24] [Impact Index Per Article: 8.0] [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
|
24
|
Bich Hanh BT, Quang NT, Park Y, Heo BE, Jeon S, Park JW, Jang J. Omadacycline Potentiates Clarithromycin Activity Against Mycobacterium abscessus. Front Pharmacol 2021; 12:790767. [PMID: 34955859 PMCID: PMC8693020 DOI: 10.3389/fphar.2021.790767] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/15/2021] [Indexed: 11/29/2022] Open
Abstract
Mycobacterium abscessus is a difficult respiratory pathogen to treat, when compared to other nontuberculus mycobacteria (NTM), due to its drug resistance. In this study, we aimed to find a new clarithromycin partner that potentiated strong, positive, synergy against M. abscessus among current anti-M. abscessus drugs, including omadacycline, amikacin, rifabutin, bedaquiline, and cefoxitine. First, we determined the minimum inhibitory concentrations required of all the drugs tested for M. abscessus subsp. abscessus CIP104536T treatment using a resazurin microplate assay. Next, the best synergistic partner for clarithromycin against M. abscessus was determined using an in vitro checkerboard combination assay. Among the drug combinations evaluated, omadacycline showed the best synergistic effect with clarithromycin, with a fractional inhibitory concentration index of 0.4. This positive effect was also observed against M. abscessus clinical isolates and anti-M. abscessus drug resistant strains. Lastly, this combination was further validated using a M. abscessus infected zebrafish model. In this model, the clarithromycin-omadacyline regimen was found to inhibit the dissemination of M. abscessus, and it significantly extended the lifespan of the M. abscessus infected zebrafish. In summation, the synergy between two anti-M. abscessus compounds, clarithromycin and omadacycline, provides an attractive foundation for a new M. abscessus treatment regimen.
Collapse
Affiliation(s)
- Bui Thi Bich Hanh
- Division of Applied Life Science (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Nguyen Thanh Quang
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Yujin Park
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Bo Eun Heo
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Seunghyeon Jeon
- Division of Life Science, Gyeongsang National University, Jinju, South Korea
| | - June-Woo Park
- Department of Environmental Toxicology and Chemistry, Korea Institute of Toxicology, Korea & Human and Environmental Toxicology Program, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Jichan Jang
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea.,Division of Life Science, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
25
|
Fujiwara K, Uesugi F, Furuuchi K, Tanaka Y, Yoshiyama T, Saotome M, Ohta K, Mitarai S, Morimoto K. Minimum Inhibitory Concentrations before and after Antibacterial Treatment in Patients with Mycobacterium abscessus Pulmonary Disease. Microbiol Spectr 2021; 9:e0192821. [PMID: 34878300 PMCID: PMC8653840 DOI: 10.1128/spectrum.01928-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 11/20/2022] Open
Abstract
The clinical importance of Mycobacterium abscessus (MABS) pulmonary disease has been increasing. However, there is still a lack of information about MIC distribution patterns and changes in clinical practice settings. The MIC results of rapidly growing mycobacteria isolated from 92 patients with nontuberculous mycobacterial pulmonary disease diagnosed from May 2019 to March 2021 were retrospectively analyzed. Most of the patients (86 patients; 93.5%) were infected with MABS; 46 with Mycobacterium abscessus subsp. abscessus (Mab), and 40 with Mycobacterium abscessus subsp. massiliense (Mma). Significant differences in susceptibility to clarithromycin (15.2% versus 80.0%, P < 0.001) and azithromycin (8.7% versus 62.5%, P < 0.001) were observed between Mab and Mma. Most isolates were susceptible to amikacin (80; 93.0%), and over half were susceptible to linezolid (48; 55.8%). Only one-quarter of isolates (22, 25.6%) were susceptible to imipenem, while more than half (56; 65.1%) had intermediate susceptibility. Fifty-one isolates (59.3%) had MIC values of less than 1 μg/mL for sitafloxacin, which were significantly higher than isolates for moxifloxacin (5; 5.8%), especially in Mab. Sixty-five (75.6%) isolates had MICs of less than 0.5 μg/mL to clofazimine. Two patients showed obvious MIC result changes: from susceptible to resistant to clarithromycin and from resistant to susceptible to amikacin and imipenem. In conclusion, MABS isolates were relatively susceptible to amikacin and linezolid, and clarithromycin and azithromycin were especially effective against Mma. In addition, sitafloxacin and clofazimine had low MICs and might be effective treatment agents. IMPORTANCE The MICs of isolates from 86 patients with Mycobacterium abscessus (MABS); 46 with Mycobacterium abscessus subsp. abscessus (Mab), and 40 with Mycobacterium abscessus subsp. massiliense (Mma) were retrospectively analyzed. The main findings are as follows: (i) Mma were significantly more susceptible to clarithromycin and azithromycin than Mab, and both subspecies tended to be more susceptible to clarithromycin than azithromycin. (ii) Most isolates were susceptible to amikacin (93.0%), and over half to linezolid (55.8%). (iii) Fifty-one isolates (59.3%) had MIC values of less than 1 μg/mL for sitafloxacin, and 65 (75.6%) had less than 0.5 μg/mL for clofazimine, which seems worth clinical investigating. (iv) Among nine cases analyzed chronological changes, only two patients showed obvious MIC result changes even after the long-term multidrug treatment. The present study revealed MICs of MABS clinical isolates before and after treatment in clinical settings, which could help develop future MABS treatments strategies.
Collapse
Affiliation(s)
- Keiji Fujiwara
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Department of Mycobacterium Reference and Research, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Department of Basic Mycobacteriosis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Fumiko Uesugi
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Koji Furuuchi
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Department of Basic Mycobacteriosis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yoshiaki Tanaka
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Takashi Yoshiyama
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Mikio Saotome
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Ken Ohta
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Satoshi Mitarai
- Department of Mycobacterium Reference and Research, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Department of Basic Mycobacteriosis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kozo Morimoto
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Division of Clinical Research, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| |
Collapse
|
26
|
Pont S, Blanc-Potard AB. Zebrafish Embryo Infection Model to Investigate Pseudomonas aeruginosa Interaction With Innate Immunity and Validate New Therapeutics. Front Cell Infect Microbiol 2021; 11:745851. [PMID: 34660345 PMCID: PMC8515127 DOI: 10.3389/fcimb.2021.745851] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/08/2021] [Indexed: 12/26/2022] Open
Abstract
The opportunistic human pathogen Pseudomonas aeruginosa is responsible for a variety of acute infections and is a major cause of mortality in chronically infected patients with cystic fibrosis (CF). Considering the intrinsic and acquired resistance of P. aeruginosa to currently used antibiotics, new therapeutic strategies against this pathogen are urgently needed. Whereas virulence factors of P. aeruginosa are well characterized, the interplay between P. aeruginosa and the innate immune response during infection remains unclear. Zebrafish embryo is now firmly established as a potent vertebrate model for the study of infectious human diseases, due to strong similarities of its innate immune system with that of humans and the unprecedented possibilities of non-invasive real-time imaging. This model has been successfully developed to investigate the contribution of bacterial and host factors involved in P. aeruginosa pathogenesis, as well as rapidly assess the efficacy of anti-Pseudomonas molecules. Importantly, zebrafish embryo appears as the state-of-the-art model to address in vivo the contribution of innate immunity in the outcome of P. aeruginosa infection. Of interest, is the finding that the zebrafish encodes a CFTR channel closely related to human CFTR, which allowed to develop a model to address P. aeruginosa pathogenesis, innate immune response, and treatment evaluation in a CF context.
Collapse
Affiliation(s)
- Stéphane Pont
- Laboratory of Pathogen-Host Interactions (LPHI), Université Montpellier, Montpellier, France.,CNRS, UMR5235, Montpellier, France
| | - Anne-Béatrice Blanc-Potard
- Laboratory of Pathogen-Host Interactions (LPHI), Université Montpellier, Montpellier, France.,CNRS, UMR5235, Montpellier, France
| |
Collapse
|
27
|
Sullivan JR, Lupien A, Kalthoff E, Hamela C, Taylor L, Munro KA, Schmeing TM, Kremer L, Behr MA. Efficacy of epetraborole against Mycobacterium abscessus is increased with norvaline. PLoS Pathog 2021; 17:e1009965. [PMID: 34637487 PMCID: PMC8535176 DOI: 10.1371/journal.ppat.1009965] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/22/2021] [Accepted: 09/23/2021] [Indexed: 12/16/2022] Open
Abstract
Mycobacterium abscessus is the most common rapidly growing non-tuberculous mycobacteria to cause pulmonary disease in patients with impaired lung function such as cystic fibrosis. M. abscessus displays high intrinsic resistance to common antibiotics and inducible resistance to macrolides like clarithromycin. As such, M. abscessus is clinically resistant to the entire regimen of front-line M. tuberculosis drugs, and treatment with antibiotics that do inhibit M. abscessus in the lab results in cure rates of 50% or less. Here, we identified epetraborole (EPT) from the MMV pandemic response box as an inhibitor against the essential protein leucyl-tRNA synthetase (LeuRS) in M. abscessus. EPT protected zebrafish from lethal M. abscessus infection and did not induce self-resistance nor against clarithromycin. Contrary to most antimycobacterials, the whole-cell activity of EPT was greater against M. abscessus than M. tuberculosis, but crystallographic and equilibrium binding data showed that EPT binds LeuRSMabs and LeuRSMtb with similar residues and dissociation constants. Since EPT-resistant M. abscessus mutants lost LeuRS editing activity, these mutants became susceptible to misaminoacylation with leucine mimics like the non-proteinogenic amino acid norvaline. Proteomic analysis revealed that when M. abscessus LeuRS mutants were fed norvaline, leucine residues in proteins were replaced by norvaline, inducing the unfolded protein response with temporal changes in expression of GroEL chaperonins and Clp proteases. This supports our in vitro data that supplementation of media with norvaline reduced the emergence of EPT mutants in both M. abscessus and M. tuberculosis. Furthermore, the combination of EPT and norvaline had improved in vivo efficacy compared to EPT in a murine model of M. abscessus infection. Our results emphasize the effectiveness of EPT against the clinically relevant cystic fibrosis pathogen M. abscessus, and these findings also suggest norvaline adjunct therapy with EPT could be beneficial for M. abscessus and other mycobacterial infections like tuberculosis. Current antimycobacterial drugs are inadequate to handle the increasing number of non-tuberculous mycobacteria infections that eclipse tuberculosis infections in many developed countries. Of particular importance for cystic fibrosis patients, Mycobacterium abscessus is notoriously difficult to treat where patients spend extended time on antibiotics with cure rates comparable to extreme drug resistant M. tuberculosis. Here, we identified epetraborole (EPT) with in vitro and in vivo activities against M. abscessus. We showed that EPT targets the editing domain of the leucyl-tRNA synthetase (LeuRS) and that escape mutants lost LeuRS editing activity, making these mutants susceptible to misaminoacylation with leucine mimics. Most importantly, combination therapy of EPT and norvaline limited the rate of EPT resistance in both M. abscessus and M. tuberculosis, and this was the first study to demonstrate improved in vivo efficacy of EPT and norvaline compared to EPT in a murine model of M. abscessus pulmonary infection. The demonstration of norvaline adjunct therapy with EPT for M. abscessus infections is promising for cystic fibrosis patients and could translate to other mycobacterial infections, such as tuberculosis.
Collapse
Affiliation(s)
- Jaryd R. Sullivan
- Department of Microbiology & Immunology, McGill University, Montréal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, Canada
- McGill International TB Centre, Montréal, Canada
| | - Andréanne Lupien
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, Canada
- McGill International TB Centre, Montréal, Canada
| | - Elias Kalthoff
- Department of Biochemistry, McGill University, Montréal, Canada
- Centre de Recherche en Biologie Structural, McGill University, Montréal, Canada
| | - Claire Hamela
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | - Lorne Taylor
- Clinical Proteomics Platform, Research Institute of the McGill University Health Centre, Montréal, Canada
| | - Kim A. Munro
- Department of Biochemistry, McGill University, Montréal, Canada
- Centre de Recherche en Biologie Structural, McGill University, Montréal, Canada
| | - T. Martin Schmeing
- Department of Biochemistry, McGill University, Montréal, Canada
- Centre de Recherche en Biologie Structural, McGill University, Montréal, Canada
| | - 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
| | - Marcel A. Behr
- Department of Microbiology & Immunology, McGill University, Montréal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, Canada
- McGill International TB Centre, Montréal, Canada
- Department of Medicine, McGill University Health Centre, Montréal, Canada
- * E-mail:
| |
Collapse
|
28
|
Quang NT, Jang J. Current Molecular Therapeutic Agents and Drug Candidates for Mycobacterium abscessus. Front Pharmacol 2021; 12:724725. [PMID: 34526902 PMCID: PMC8435730 DOI: 10.3389/fphar.2021.724725] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022] Open
Abstract
Mycobacterium abscessus has been recognised as a dreadful respiratory pathogen among the non-tuberculous mycobacteria (NTM) because of misdiagnosis, prolonged therapy with poor treatment outcomes and a high cost. This pathogen also shows extremely high antimicrobial resistance against current antibiotics, including the anti-tuberculosis agents. Therefore, current chemotherapies require a long curative period and the clinical outcomes are not satisfactory. Thus, there is an urgent need for discovering and developing novel, more effective anti-M. abscessus drugs. In this review, we sum the effectiveness of the current anti-M. abscessus drugs and drug candidates. Furthermore, we describe the shortcomings and difficulties associated with M. abscessus drug discovery and development.
Collapse
Affiliation(s)
- Nguyen Thanh Quang
- Molecular Mechanisms of Antibiotics, Division of Life Science, Department of Bio and Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Jichan Jang
- Molecular Mechanisms of Antibiotics, Division of Life Science, Department of Bio and Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
29
|
Johansen MD, Alcaraz M, Dedrick RM, Roquet-Banères F, Hamela C, Hatfull GF, Kremer L. Mycobacteriophage-antibiotic therapy promotes enhanced clearance of drug-resistant Mycobacterium abscessus. Dis Model Mech 2021; 14:272140. [PMID: 34530447 PMCID: PMC8461822 DOI: 10.1242/dmm.049159] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/26/2021] [Indexed: 12/16/2022] Open
Abstract
Infection by multidrug-resistant Mycobacterium abscessus is increasingly prevalent in cystic fibrosis (CF) patients, leaving clinicians with few therapeutic options. A compassionate study showed the clinical improvement of a CF patient with a disseminated M. abscessus (GD01) infection, following injection of a phage cocktail, including phage Muddy. Broadening the use of phage therapy in patients as a potential antibacterial alternative necessitates the development of biological models to improve the reliability and successful prediction of phage therapy in the clinic. Herein, we demonstrate that Muddy very efficiently lyses GD01 in vitro, an effect substantially increased with standard drugs. Remarkably, this cooperative activity was retained in an M. abscessus model of infection in CFTR-depleted zebrafish, associated with a striking increase in larval survival and reduction in pathological signs. The activity of Muddy was lost in macrophage-ablated larvae, suggesting that successful phage therapy relies on functional innate immunity. CFTR-depleted zebrafish represent a practical model to rapidly assess phage treatment efficacy against M. abscessus isolates, allowing the identification of drug combinations accompanying phage therapy and treatment prediction in patients. This article has an associated First Person interview with the first author of the paper. Summary: A zebrafish model of infection was developed to evaluate the in vivo cooperative activity of specific phages and antibiotics for the treatment of Mycobacterium abscessus infection.
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 34293, France
| | - Matthéo Alcaraz
- Institut de Recherche en Infectiologie de Montpellier, Centre National de la Recherche Scientifique UMR 9004, Université de Montpellier, Montpellier 34293, France
| | - Rebekah M Dedrick
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Françoise Roquet-Banères
- Institut de Recherche en Infectiologie de Montpellier, Centre National de la Recherche Scientifique UMR 9004, Université de Montpellier, Montpellier 34293, France
| | - Claire Hamela
- Institut de Recherche en Infectiologie de Montpellier, Centre National de la Recherche Scientifique UMR 9004, Université de Montpellier, Montpellier 34293, France
| | - Graham F Hatfull
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Laurent Kremer
- Institut de Recherche en Infectiologie de Montpellier, Centre National de la Recherche Scientifique UMR 9004, Université de Montpellier, Montpellier 34293, France.,INSERM, Institut de Recherche en Infectiologie de Montpellier, Montpellier 34293, France
| |
Collapse
|
30
|
Kim T, Hanh BTB, Heo B, Quang N, Park Y, Shin J, Jeon S, Park JW, Samby K, Jang J. A Screening of the MMV Pandemic Response Box Reveals Epetraborole as a New Potent Inhibitor against Mycobacterium abscessus. Int J Mol Sci 2021; 22:ijms22115936. [PMID: 34073006 PMCID: PMC8199016 DOI: 10.3390/ijms22115936] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022] Open
Abstract
Mycobacterium abscessus is the one of the most feared bacterial respiratory pathogens in the world. Unfortunately, there are many problems with the current M. abscessus therapies available. These problems include misdiagnoses, high drug resistance, poor long-term treatment outcomes, and high costs. Until now, there have only been a few new compounds or drug formulations which are active against M. abscessus, and these are present in preclinical and clinical development only. With that in mind, new and more powerful anti-M. abscessus medicines need to be discovered and developed. In this study, we conducted an in vitro-dual screen against M. abscessus rough (R) and smooth (S) variants using a Pandemic Response Box and identified epetraborole as a new effective candidate for M. abscessus therapy. For further validation, epetraborole showed significant activity against the growth of the M. abscessus wild-type strain, three subspecies, drug-resistant strains and clinical isolates in vitro, while also inhibiting the growth of M. abscessus that reside in macrophages without cytotoxicity. Furthermore, the in vivo efficacy of epetraborole in the zebrafish infection model was greater than that of tigecycline. Thus, we concluded that epetraborole is a potential anti-M. abscessus candidate in the M. abscessus drug search.
Collapse
Affiliation(s)
- Taeho Kim
- Division of Applied Life Science (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (T.K.); (B.-T.-B.H.)
| | - Bui-Thi-Bich Hanh
- Division of Applied Life Science (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (T.K.); (B.-T.-B.H.)
| | - Boeun Heo
- Molecular Mechanisms of Antibiotics, Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (B.H.); (N.Q.); (Y.P.); (J.S.)
| | - Nguyenthanh Quang
- Molecular Mechanisms of Antibiotics, Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (B.H.); (N.Q.); (Y.P.); (J.S.)
| | - Yujin Park
- Molecular Mechanisms of Antibiotics, Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (B.H.); (N.Q.); (Y.P.); (J.S.)
| | - Jihyeon Shin
- Molecular Mechanisms of Antibiotics, Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (B.H.); (N.Q.); (Y.P.); (J.S.)
| | - Seunghyeon Jeon
- Division of Life Science, Gyeongsang National University, Jinju 52828, Korea;
| | - June-Woo Park
- Department of Environmental Toxicology and Chemistry, Korea Institute of Toxicology, Jinju 52843, Korea;
- Human and Environmental Toxicology Program, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Kirandeep Samby
- Medicines for Malaria Venture (MMV), 20, Route de Pré-Bois, 1215 Geneva, Switzerland;
| | - Jichan Jang
- Molecular Mechanisms of Antibiotics, Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (B.H.); (N.Q.); (Y.P.); (J.S.)
- Correspondence: ; Tel.: +82-055-772-1368
| |
Collapse
|
31
|
A Novel Infection Protocol in Zebrafish Embryo to Assess Pseudomonas aeruginosa Virulence and Validate Efficacy of a Quorum Sensing Inhibitor In Vivo. Pathogens 2021; 10:pathogens10040401. [PMID: 33805384 PMCID: PMC8065929 DOI: 10.3390/pathogens10040401] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/23/2022] Open
Abstract
The opportunistic human pathogen Pseudomonas aeruginosa is responsible for a variety of acute infections and is a major cause of mortality in chronically infected cystic fibrosis patients. Due to increased resistance to antibiotics, new therapeutic strategies against P. aeruginosa are urgently needed. In this context, we aimed to develop a simple vertebrate animal model to rapidly assess in vivo drug efficacy against P. aeruginosa. Zebrafish are increasingly considered for modeling human infections caused by bacterial pathogens, which are commonly microinjected in embryos. In the present study, we established a novel protocol for zebrafish infection by P. aeruginosa based on bath immersion in 96-well plates of tail-injured embryos. The immersion method, followed by a 48-hour survey of embryo viability, was first validated to assess the virulence of P. aeruginosa wild-type PAO1 and a known attenuated mutant. We then validated its relevance for antipseudomonal drug testing by first using a clinically used antibiotic, ciprofloxacin. Secondly, we used a novel quorum sensing (QS) inhibitory molecule, N-(2-pyrimidyl)butanamide (C11), the activity of which had been validated in vitro but not previously tested in any animal model. A significant protective effect of C11 was observed on infected embryos, supporting the ability of C11 to attenuate in vivo P. aeruginosa pathogenicity. In conclusion, we present here a new and reliable method to compare the virulence of P. aeruginosa strains in vivo and to rapidly assess the efficacy of clinically relevant drugs against P. aeruginosa, including new antivirulence compounds.
Collapse
|
32
|
Saxena S, Spaink HP, Forn-Cuní G. Drug Resistance in Nontuberculous Mycobacteria: Mechanisms and Models. BIOLOGY 2021; 10:biology10020096. [PMID: 33573039 PMCID: PMC7911849 DOI: 10.3390/biology10020096] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 02/07/2023]
Abstract
The genus Mycobacteria comprises a multitude of species known to cause serious disease in humans, including Mycobacterium tuberculosis and M. leprae, the responsible agents for tuberculosis and leprosy, respectively. In addition, there is a worldwide spike in the number of infections caused by a mixed group of species such as the M. avium, M. abscessus and M. ulcerans complexes, collectively called nontuberculous mycobacteria (NTMs). The situation is forecasted to worsen because, like tuberculosis, NTMs either naturally possess or are developing high resistance against conventional antibiotics. It is, therefore, important to implement and develop models that allow us to effectively examine the fundamental questions of NTM virulence, as well as to apply them for the discovery of new and improved therapies. This literature review will focus on the known molecular mechanisms behind drug resistance in NTM and the current models that may be used to test new effective antimicrobial therapies.
Collapse
|
33
|
Rifabutin Is Bactericidal against Intracellular and Extracellular Forms of Mycobacterium abscessus. Antimicrob Agents Chemother 2020; 64:AAC.00363-20. [PMID: 32816730 DOI: 10.1128/aac.00363-20] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 08/03/2020] [Indexed: 12/26/2022] Open
Abstract
Mycobacterium abscessus is increasingly recognized as an emerging opportunistic pathogen causing severe lung diseases. As it is intrinsically resistant to most conventional antibiotics, there is an unmet medical need for effective treatments. Repurposing of clinically validated pharmaceuticals represents an attractive option for the development of chemotherapeutic alternatives against M. abscessus infections. In this context, rifabutin (RFB) has been shown to be active against M. abscessus and has raised renewed interest in using rifamycins for the treatment of M. abscessus pulmonary diseases. Here, we compared the in vitro and in vivo activity of RFB against the smooth and rough variants of M. abscessus, differing in their susceptibility profiles to several drugs and physiopathologial characteristics. While the activity of RFB is greater against rough strains than in smooth strains in vitro, suggesting a role of the glycopeptidolipid layer in susceptibility to RFB, both variants were equally susceptible to RFB inside human macrophages. RFB treatment also led to a reduction in the number and size of intracellular and extracellular mycobacterial cords. Furthermore, RFB was highly effective in a zebrafish model of infection and protected the infected larvae from M. abscessus-induced killing. This was corroborated by a significant reduction in the overall bacterial burden, as well as decreased numbers of abscesses and cords, two major pathophysiological traits in infected zebrafish. This study indicates that RFB is active against M. abscessus both in vitro and in vivo, further supporting its potential usefulness as part of combination regimens targeting this difficult-to-treat mycobacterium.
Collapse
|
34
|
Brzostek J, Fatin A, Chua WH, Tan HY, Dick T, Gascoigne NRJ. Single Cell Analysis of Drug Susceptibility of Mycobacterium Abscessus During Macrophage Infection. Antibiotics (Basel) 2020; 9:antibiotics9100711. [PMID: 33080864 PMCID: PMC7650608 DOI: 10.3390/antibiotics9100711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 11/16/2022] Open
Abstract
Mycobacterium abscessus is an emerging health risk to immunocompromised individuals and to people with pre-existing pulmonary conditions. As M. abscessus possesses multiple mechanisms of drug resistance, treatments of M. abscessus are of poor efficacy. Therefore, there is an urgent need for new therapeutic strategies targeting M. abscessus. We describe an experimental system for screening of compounds for their antimicrobial activity against intracellular M. abscessus using flow cytometry and imaging flow cytometry. The assay allows simultaneous analysis of multiple parameters, such as proportion of infected host cells, bacterial load per host cell from the infected population, and host cell viability. We verified the suitability of this method using two antibiotics with known activity against M. abscessus: clarithromycin and amikacin. Our analysis revealed a high degree of infection heterogeneity, which correlated with host cell size. A higher proportion of the larger host cells is infected with M. abscessus as compared to smaller host cells, and infected larger cells have higher intracellular bacterial burden than infected smaller cells. Clarithromycin treatment has a more pronounced effect on smaller host cells than on bigger host cells, suggesting that heterogeneity within the host cell population has an effect on antibiotic susceptibility of intracellular bacteria.
Collapse
Affiliation(s)
- Joanna Brzostek
- Department of Microbiology and Immunology, Yong Loo Lin School of Medcine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore; (A.F.); (W.H.C.); (H.Y.T.); (T.D.)
- Correspondence: (J.B.); (N.R.J.G.)
| | - Amierah Fatin
- Department of Microbiology and Immunology, Yong Loo Lin School of Medcine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore; (A.F.); (W.H.C.); (H.Y.T.); (T.D.)
| | - Wen Hui Chua
- Department of Microbiology and Immunology, Yong Loo Lin School of Medcine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore; (A.F.); (W.H.C.); (H.Y.T.); (T.D.)
| | - Hui Yi Tan
- Department of Microbiology and Immunology, Yong Loo Lin School of Medcine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore; (A.F.); (W.H.C.); (H.Y.T.); (T.D.)
| | - Thomas Dick
- Department of Microbiology and Immunology, Yong Loo Lin School of Medcine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore; (A.F.); (W.H.C.); (H.Y.T.); (T.D.)
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine at Seton Hall University, Nutley, NJ 07110, USA
| | - Nicholas R. J. Gascoigne
- Department of Microbiology and Immunology, Yong Loo Lin School of Medcine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore; (A.F.); (W.H.C.); (H.Y.T.); (T.D.)
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117545, Singapore
- Correspondence: (J.B.); (N.R.J.G.)
| |
Collapse
|
35
|
Le Moigne V, Roux AL, Jobart-Malfait A, Blanc L, Chaoui K, Burlet-Schiltz O, Gaillard JL, Canaan S, Nigou J, Herrmann JL. A TLR2-Activating Fraction From Mycobacterium abscessus Rough Variant Demonstrates Vaccine and Diagnostic Potential. Front Cell Infect Microbiol 2020; 10:432. [PMID: 32984067 PMCID: PMC7481331 DOI: 10.3389/fcimb.2020.00432] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 07/14/2020] [Indexed: 11/17/2022] Open
Abstract
Mycobacterium abscessus is a prevalent pathogenic mycobacterium in cystic fibrosis (CF) patients and one of the most highly drug resistant mycobacterial species to antimicrobial agents. It possesses the property to transition from a smooth (S) to a rough (R) morphotype, thereby influencing the host innate immune response. This transition from the S to the R morphotype takes place in patients with an exacerbation of the disease and a persistence of M. abscessus. We have previously shown that the exacerbation of the Toll-like receptor 2 (TLR2)-mediated inflammatory response, following this S to R transition, is essentially due to overproduction of bacilli cell envelope surface compounds, which we were able to extract by mechanical treatment and isolation by solvent partition in a fraction called interphase. Here, we set up a purification procedure guided by bioactivity to isolate a fraction from the R variant of M. abscessus cells which exhibits a high TLR2 stimulating activity, referred to as TLR2-enriched fraction (TLR2eF). As expected, TLR2eF was found to contain several lipoproteins and proteins known to be stimuli for TLR2. Vaccination with TLR2eF showed no protection toward an M. abscessus aerosol challenge, but provided mild protection in ΔF508 mice and their FVB littermates when intravenously challenged by M. abscessus. Interestingly however, antibodies against TLR2eF compounds were detected during disease in CF patients. In conclusion, we show the potential for compounds in TLR2eF as vaccine and diagnostic candidates, in order to enhance diagnosis, prevent and/or treat M. abscessus-related infections.
Collapse
Affiliation(s)
- Vincent Le Moigne
- Université Paris-Saclay, UVSQ, Inserm, Infection et inflammation, Montigny-le-Bretonneux, France
| | - Anne-Laure Roux
- Université Paris-Saclay, UVSQ, Inserm, Infection et inflammation, Montigny-le-Bretonneux, France
| | - Aude Jobart-Malfait
- Université Paris-Saclay, UVSQ, Inserm, Infection et inflammation, Montigny-le-Bretonneux, France
| | - Landry Blanc
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, Toulouse, France
| | - Karima Chaoui
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, Toulouse, France
| | - Jean-Louis Gaillard
- Université Paris-Saclay, UVSQ, Inserm, Infection et inflammation, Montigny-le-Bretonneux, France
| | - Stéphane Canaan
- Université Aix-Marseille, CNRS, LISM, IMM FR3479, Marseille, France
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, Toulouse, France
| | - Jean-Louis Herrmann
- Université Paris-Saclay, UVSQ, Inserm, Infection et inflammation, Montigny-le-Bretonneux, France.,APHP, GHU Paris-Saclay, Hôpital Raymond Poincaré, Service de Microbiologie, Garches, France
| |
Collapse
|
36
|
Shapira T, Rankine-Wilson L, Chao JD, Pichler V, Rens C, Pfeifer T, Av-Gay Y. High-Content Screening of Eukaryotic Kinase Inhibitors Identify CHK2 Inhibitor Activity Against Mycobacterium tuberculosis. Front Microbiol 2020; 11:553962. [PMID: 33042061 PMCID: PMC7530171 DOI: 10.3389/fmicb.2020.553962] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022] Open
Abstract
A screen of a eukaryotic kinase inhibitor library in an established intracellular infection model identified a set of drug candidates enabling intracellular killing of Mycobacterium tuberculosis (M.tb). Screen validity was confirmed internally by a Z′ = 0.5 and externally by detecting previously reported host-targeting anti-M.tb compounds. Inhibitors of the CHK kinase family, specifically checkpoint kinase 2 (CHK2), showed the highest inhibition and lowest toxicity of all kinase families. The screen identified and validated DDUG, a CHK2 inhibitor, as a novel bactericidal anti-M.tb compound. CHK2 inhibition by RNAi phenocopied the intracellular inhibitory effect of DDUG. DDUG was active intracellularly against M.tb, but not other mycobacteria. DDUG also had extracellular activity against 4 of 12 bacteria tested, including M.tb. Combined, these observations suggest DDUG acts in tandem against both host and pathogen. Importantly, DDUG’s validation highlights the screening and analysis methodology developed for this screen, which identified novel host-directed anti-M.tb compounds.
Collapse
Affiliation(s)
- Tirosh Shapira
- Division of Infectious Diseases, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Leah Rankine-Wilson
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Joseph D Chao
- Division of Infectious Diseases, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Virginia Pichler
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Celine Rens
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Tom Pfeifer
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Yossef Av-Gay
- Division of Infectious Diseases, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
37
|
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
|
38
|
Johansen MD, Kremer L. CFTR Depletion Confers Hypersusceptibility to Mycobacterium fortuitum in a Zebrafish Model. Front Cell Infect Microbiol 2020; 10:357. [PMID: 32850470 PMCID: PMC7396536 DOI: 10.3389/fcimb.2020.00357] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/10/2020] [Indexed: 12/15/2022] Open
Abstract
The Mycobacterium fortuitum complex comprises several closely related species, causing pulmonary and extra-pulmonary infections. However, there is very limited knowledge about the disease pathogenesis involved in M. fortuitum infections, particularly due to the lack of suitable animal models. Using the zebrafish model, we show that embryos are susceptible to M. fortuitum infection in a dose-dependent manner. Furthermore, zebrafish embryos form granulomas from as early as 2 days post-infection, recapitulating critical aspects of mycobacterial pathogenesis observed in other pathogenic species. The formation of extracellular cords in infected embryos highlights a previously unknown pathogenic feature of M. fortuitum. The formation of large corded structures occurs also during in vitro growth, suggesting that this is not a host-adapted stress mechanism deployed during infection. Moreover, transient macrophage depletion led to rapid embryo death with increased extracellular cords, indicating that macrophages are essential determinants of M. fortuitum infection control. Importantly, morpholino depletion of the cystic fibrosis transmembrane conductance regulator (cftr) significantly increased embryo death, bacterial burden, bacterial cords and abscesses. There was a noticeable decrease in the number of cftr-deficient infected embryos with granulomas as compared to infected controls, suggesting that loss of CFTR leads to impaired host immune responses and confers hypersusceptiblity to M. fortuitum infection. Overall, these findings highlight the application of the zebrafish embryo to study M. fortuitum and emphasizes previously unexplored aspects of disease pathogenesis of this significant mycobacterial species.
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
| | - 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
|
39
|
Efficacy of Bedaquiline, Alone or in Combination with Imipenem, against Mycobacterium abscessus in C3HeB/FeJ Mice. Antimicrob Agents Chemother 2020; 64:AAC.00114-20. [PMID: 32253217 DOI: 10.1128/aac.00114-20] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/26/2020] [Indexed: 12/14/2022] Open
Abstract
Mycobacterium abscessus lung infections remain difficult to treat. Recent studies have recognized the power of new combinations of antibiotics, such as bedaquiline and imipenem, although in vitro data have questioned this combination. We report that the efficacy of bedaquiline-imipenem combination treatment relies essentially on the activity of bedaquiline in a C3HeB/FeJ mice model of infection with a rough variant of M. abscessus The addition of imipenem contributed to clearing the infection in the spleen.
Collapse
|
40
|
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: 449] [Impact Index Per Article: 89.8] [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
|
41
|
Raynaud C, Daher W, Johansen MD, Roquet-Banères F, Blaise M, Onajole OK, Kozikowski AP, Herrmann JL, Dziadek J, Gobis K, Kremer L. Active Benzimidazole Derivatives Targeting the MmpL3 Transporter in Mycobacterium abscessus. ACS Infect Dis 2020; 6:324-337. [PMID: 31860799 DOI: 10.1021/acsinfecdis.9b00389] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The prevalence of pulmonary infections due to nontuberculous mycobacteria such as Mycobacterium abscessus has been increasing and surpassing tuberculosis (TB) in some industrialized countries. Because of intrinsic resistance to most antibiotics that drastically limits conventional chemotherapeutic treatment options, new anti-M. abscessus therapeutics are urgently needed against this emerging pathogen. Extensive screening of a library of benzimidazole derivatives that were previously shown to be active against Mycobacterium tuberculosis led to the identification of a lead compound exhibiting very potent in vitro activity against a wide panel of M. abscessus clinical strains. Designated EJMCh-6, this compound, a 2-(2-cyclohexylethyl)-5,6-dimethyl-1H-benzo[d]imidazole), also exerted very strong activity against intramacrophage-residing M. abscessus. Moreover, the treatment of infected zebrafish embryos with EJMCh-6 was correlated with significantly increased embryo survival and a decrease in the bacterial burden as compared to those for untreated fish. Insights into the mechanism of action were inferred from the generation of spontaneous benzimidazole-resistant strains and the identification of a large set of missense mutations in MmpL3, the mycolic acid transporter in mycobacteria. Overexpression of the mutated mmpL3 alleles in a susceptible M. abscessus strain was associated with high resistance levels to EJMCh-6 and to other known MmpL3 inhibitors. Mapping the mutations conferring resistance on an MmpL3 three-dimensional homology model defined a potential EJMCh-6-binding cavity. These data emphasize a yet unexploited chemical structure class against M. abscessus with promising translational development for the treatment of M. abscessus lung diseases.
Collapse
Affiliation(s)
- Clément Raynaud
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France
| | - Wassim Daher
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France
| | - Matt D. Johansen
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France
| | - Françoise Roquet-Banères
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France
| | - Mickael Blaise
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France
| | - Oluseye K. Onajole
- Department of Biological, Physical and Health Sciences, Roosevelt University, 425 S. Wabash Avenue, Chicago, Illinois 60605, United States
| | - Alan P. Kozikowski
- StarWise Therapeutics LLC, 2020 N. Lincoln Park West, Chicago, Illinois 60614, United States
| | - Jean-Louis Herrmann
- 2I, UVSQ, INSERM UMR1173, Université Paris-Saclay, 2 avenue de la Source de la Bièvre, 78180 Montigny-Le-Bretonneux, France
- APHP, GHU-Paris Saclay, Hôpital Raymond Poincaré, Garches, France
| | - Jaroslaw Dziadek
- Institute for Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź 93-232, Poland
| | - Katarzyna Gobis
- Department of Organic Chemistry, Medical University of Gdansk, 107 Gen. Hallera Avenue, 80-416 Gdansk, Poland
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293 Montpellier, France
- INSERM, IRIM, 34293 Montpellier, France
| |
Collapse
|
42
|
Dissecting erm(41)-Mediated Macrolide-Inducible Resistance in Mycobacterium abscessus. Antimicrob Agents Chemother 2020; 64:AAC.01879-19. [PMID: 31791943 DOI: 10.1128/aac.01879-19] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/25/2019] [Indexed: 12/18/2022] Open
Abstract
Macrolides are the cornerstone of Mycobacterium abscessus multidrug therapy, despite that most patients respond poorly to this class of antibiotics due to the inducible resistance phenotype that occurs during drug treatment. This mechanism is driven by the macrolide-inducible ribosomal methylase encoded by erm(41), whose expression is activated by the transcriptional regulator WhiB7. However, it has been debated whether clarithromycin and azithromycin differ in the extent to which they induce erm(41)-mediated macrolide resistance. Herein, we show that macrolide resistance is induced more rapidly in various M. abscessus isolates upon exposure to azithromycin than to clarithromycin, based on MIC determination. Macrolide-induced expression of erm(41) was assessed in vivo using a strain carrying tdTomato placed under the control of the erm(41) promoter. Visualization of fluorescent bacilli in infected zebrafish demonstrates that azithromycin and clarithromycin activate erm(41) expression in vivo That azithromycin induces a more rapid expression of erm(41) was confirmed by measuring the β-galactosidase activity of a reporter strain in which lacZ was placed under the control of the erm(41) promoter. Shortening the promoter region in the lacZ reporter plasmid identified DNA elements involved in the regulation of erm(41) expression, particularly an AT-rich motif sharing partial conservation with the WhiB7-binding site. Mutation of this motif abrogated the macrolide-induced and WhiB7-dependent expression of erm(41). This study provides new mechanistic information on the adaptive response to macrolide treatment in M. abscessus.
Collapse
|
43
|
THP-1 and Dictyostelium Infection Models for Screening and Characterization of Anti-Mycobacterium abscessus Hit Compounds. Antimicrob Agents Chemother 2019; 64:AAC.01601-19. [PMID: 31636068 DOI: 10.1128/aac.01601-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/10/2019] [Indexed: 12/23/2022] Open
Abstract
!!NCR1!! presents a great challenge to antimycobacterial therapy due to its innate resistance against most antibiotics. M. abscessus is able to grow intracellularly in human macrophages, suggesting that intracellular models can facilitate drug discovery. Thus, we have developed two host cell models: human macrophages for use in a new high-content screening method for M. abscessus growth and a Dictyostelium discoideum infection model with the potential to simplify downstream genetic analysis of host cell factors. A screen of 568 antibiotics for activity against intracellular M. abscessus led to the identification of two hit compounds with distinct growth inhibition. A collection of 317 human kinase inhibitors was analyzed, with the results yielding three compounds with an inhibitory effect on mycobacterial growth, strengthening the notion that host-directed therapy can be applied for M. abscessus.
Collapse
|
44
|
Kim TH, Hanh BTB, Kim G, Lee DG, Park JW, Lee SE, Kim JS, Kim BS, Ryoo S, Jo EK, Jang J. Thiostrepton: A Novel Therapeutic Drug Candidate for Mycobacterium abscessus Infection. Molecules 2019; 24:molecules24244511. [PMID: 31835481 PMCID: PMC6943738 DOI: 10.3390/molecules24244511] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/06/2019] [Accepted: 12/06/2019] [Indexed: 02/03/2023] Open
Abstract
Mycobacterium abscessus is a rapid-growing, multidrug-resistant, non-tuberculous mycobacterial species responsible for a variety of human infections, such as cutaneous and pulmonary infections. M. abscessus infections are very difficult to eradicate due to the natural and acquired multidrug resistance profiles of M. abscessus. Thus, there is an urgent need for the development of effective drugs or regimens against M. abscessus infections. Here, we report the activity of a US Food and Drug Administration approved drug, thiostrepton, against M. abscessus. We found that thiostrepton significantly inhibited the growth of M. abscessus wild-type strains, subspecies, clinical isolates, and drug-resistant mutants in vitro and in macrophages. In addition, treatment of macrophages with thiostrepton significantly decreased proinflammatory cytokine production in a dose-dependent manner, suggesting an inhibitory effect of thiostrepton on inflammation induced during M. abscessus infection. We further showed that thiostrepton exhibits antimicrobial effects in vivo using a zebrafish model of M. abscessus infection.
Collapse
Affiliation(s)
- Tae Ho Kim
- Molecular Mechanisms of Antibiotics, Division of Life Science, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
- Division of Applied Life Science (BK21plus Program), Gyeongsang National University, Jinju 52828, Korea
| | - Bui Thi Bich Hanh
- Molecular Mechanisms of Antibiotics, Division of Life Science, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
- Division of Applied Life Science (BK21plus Program), Gyeongsang National University, Jinju 52828, Korea
| | - Guehye Kim
- Molecular Mechanisms of Antibiotics, Division of Life Science, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
- Division of Applied Life Science (BK21plus Program), Gyeongsang National University, Jinju 52828, Korea
| | - Da-Gyum Lee
- Molecular Mechanisms of Antibiotics, Division of Life Science, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
- Clinical Research Centre, Masan National Tuberculosis Hospital, Changwon 51755, Korea
| | - June-Woo Park
- Future Environmental Research Center, Korea Institute of Toxicology, Jinju 52834, Korea
- Human and Environmental Toxicology Program, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - So Eui Lee
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea (E.-K.J.)
- Infection Control Convergence Research Center, Chungnam National University, Daejeon 35015, Korea
| | - Jae-Sung Kim
- Department of Bionano Technology, Hanyang University, Seoul 04763, Korea
| | - Byoung Soo Kim
- Department of Radiopharmaceutical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea
| | - Sungweon Ryoo
- Clinical Research Centre, Masan National Tuberculosis Hospital, Changwon 51755, Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea (E.-K.J.)
- Infection Control Convergence Research Center, Chungnam National University, Daejeon 35015, Korea
| | - Jichan Jang
- Molecular Mechanisms of Antibiotics, Division of Life Science, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
- Division of Applied Life Science (BK21plus Program), Gyeongsang National University, Jinju 52828, Korea
- Correspondence: ; Tel.: +82-553-772-1368
| |
Collapse
|
45
|
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: 23] [Impact Index Per Article: 3.8] [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
|
46
|
Le Moigne V, Bernut A, Cortès M, Viljoen A, Dupont C, Pawlik A, Gaillard JL, Misguich F, Crémazy F, Kremer L, Herrmann JL. Lsr2 Is an Important Determinant of Intracellular Growth and Virulence in Mycobacterium abscessus. Front Microbiol 2019; 10:905. [PMID: 31114557 PMCID: PMC6503116 DOI: 10.3389/fmicb.2019.00905] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 04/09/2019] [Indexed: 12/30/2022] Open
Abstract
Mycobacterium abscessus, a pathogen responsible for severe lung infections in cystic fibrosis patients, exhibits either smooth (S) or rough (R) morphotypes. The S-to-R transition correlates with inhibition of the synthesis and/or transport of glycopeptidolipids (GPLs) and is associated with an increase of pathogenicity in animal and human hosts. Lsr2 is a small nucleoid-associated protein highly conserved in mycobacteria, including M. abscessus, and is a functional homolog of the heat-stable nucleoid-structuring protein (H-NS). It is essential in Mycobacterium tuberculosis but not in the non-pathogenic model organism Mycobacterium smegmatis. It acts as a master transcriptional regulator of multiple genes involved in virulence and immunogenicity through binding to AT-rich genomic regions. Previous transcriptomic studies, confirmed here by quantitative PCR, showed increased expression of lsr2 (MAB_0545) in R morphotypes when compared to their S counterparts, suggesting a possible role of this protein in the virulence of the R form. This was addressed by generating lsr2 knock-out mutants in both S (Δlsr2-S) and R (Δlsr2-R) variants, demonstrating that this gene is dispensable for M. abscessus growth. We show that the wild-type S variant, Δlsr2-S and Δlsr2-R strains were more sensitive to H2O2 as compared to the wild-type R variant of M. abscessus. Importantly, virulence of the Lsr2 mutants was considerably diminished in cellular models (macrophage and amoeba) as well as in infected animals (mouse and zebrafish). Collectively, these results emphasize the importance of Lsr2 in M. abscessus virulence.
Collapse
Affiliation(s)
| | - Audrey Bernut
- UMR 9004, Centre National de la Recherche Scientifique, Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, Montpellier, France
| | | | - Albertus Viljoen
- UMR 9004, Centre National de la Recherche Scientifique, Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, Montpellier, France
| | - Christian Dupont
- UMR 9004, Centre National de la Recherche Scientifique, Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, Montpellier, France
| | - Alexandre Pawlik
- Unité de Pathogénomique Mycobactérienne, Institut Pasteur, Paris, France
| | - Jean-Louis Gaillard
- 2I, UVSQ, INSERM, Université Paris-Saclay, Versailles, France.,APHP, GHU PIFO, Hôpital Raymond-Poincaré - Hôpital Ambroise-Paré, Boulogne-Billancourt, France
| | | | | | - Laurent Kremer
- UMR 9004, Centre National de la Recherche Scientifique, Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, Montpellier, France.,INSERM, Institut de Recherche en Infectiologie de Montpellier, Montpellier, France
| | - Jean-Louis Herrmann
- 2I, UVSQ, INSERM, Université Paris-Saclay, Versailles, France.,APHP, GHU PIFO, Hôpital Raymond-Poincaré - Hôpital Ambroise-Paré, Boulogne-Billancourt, France
| |
Collapse
|
47
|
Baldwin SL, Larsen SE, Ordway D, Cassell G, Coler RN. The complexities and challenges of preventing and treating nontuberculous mycobacterial diseases. PLoS Negl Trop Dis 2019; 13:e0007083. [PMID: 30763316 PMCID: PMC6375572 DOI: 10.1371/journal.pntd.0007083] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Seemingly innocuous nontuberculous mycobacteria (NTM) species, classified by their slow or rapid growth rates, can cause a wide range of illnesses, from skin ulceration to severe pulmonary and disseminated disease. Despite their worldwide prevalence and significant disease burden, NTM do not garner the same financial or research focus as Mycobacterium tuberculosis. In this review, we outline the most abundant of over 170 NTM species and inadequacies of diagnostics and treatments and weigh the advantages and disadvantages of currently available in vivo animal models of NTM. In order to effectively combat this group of mycobacteria, more research focused on appropriate animal models of infection, screening of chemotherapeutic compounds, and development of anti-NTM vaccines and diagnostics is urgently needed.
Collapse
Affiliation(s)
- Susan L. Baldwin
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Sasha E. Larsen
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Diane Ordway
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Gail Cassell
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rhea N. Coler
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- PAI Life Sciences, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
48
|
Richter A, Strauch A, Chao J, Ko M, Av-Gay Y. Screening of Preselected Libraries Targeting Mycobacterium abscessus for Drug Discovery. Antimicrob Agents Chemother 2018; 62:e00828-18. [PMID: 30012760 PMCID: PMC6125491 DOI: 10.1128/aac.00828-18] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/08/2018] [Indexed: 12/23/2022] Open
Abstract
Mycobacterium abscessus is intrinsically resistant to many antimycobacterial antibiotics, which presents serious problems in therapy. Here, we describe the development of a novel phenotype-based microscopic and computerized imaging drug screening approach. A pilot screen of 568 compounds from two libraries identified 17 hits. Eleven of these compounds are described for the first time as active against M. abscessus The impact of growth media on the activity of these compounds was tested, revealing that cation-adjusted Mueller-Hinton broth (MHII) supports better growth of actively replicating M. abscessus and improves the activity of associated compounds.
Collapse
Affiliation(s)
- Adrian Richter
- Division of Infectious Diseases, Department of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Angelika Strauch
- Division of Infectious Diseases, Department of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joseph Chao
- Division of Infectious Diseases, Department of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mary Ko
- Division of Infectious Diseases, Department of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yossef Av-Gay
- Division of Infectious Diseases, Department of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
49
|
Gutiérrez AV, Viljoen A, Ghigo E, Herrmann JL, Kremer L. Glycopeptidolipids, a Double-Edged Sword of the Mycobacterium abscessus Complex. Front Microbiol 2018; 9:1145. [PMID: 29922253 PMCID: PMC5996870 DOI: 10.3389/fmicb.2018.01145] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/14/2018] [Indexed: 01/14/2023] Open
Abstract
Mycobacterium abscessus is a rapidly-growing species causing a diverse panel of clinical manifestations, ranging from cutaneous infections to severe respiratory disease. Its unique cell wall, contributing largely to drug resistance and to pathogenicity, comprises a vast panoply of complex lipids, among which the glycopeptidolipids (GPLs) have been the focus of intense research. These lipids fulfill various important functions, from sliding motility or biofilm formation to interaction with host cells and intramacrophage trafficking. Being highly immunogenic, the induction of a strong humoral response is likely to select for rough low-GPL producers. These, in contrast to the smooth high-GPL producers, display aggregative properties, which strongly impacts upon intracellular survival. A propensity to grow as extracellular cords allows these low-GPL producing bacilli to escape the innate immune defenses. Transitioning from high-GPL to low-GPL producers implicates mutations within genes involved in biosynthesis or transport of GPL. This leads to induction of an intense pro-inflammatory response and robust and lethal infections in animal models, explaining the presence of rough isolates in patients with decreased pulmonary functions. Herein, we will discuss how, thanks to the generation of defined GPL mutants and the development of appropriate cellular and animal models to study pathogenesis, GPL contribute to M. abscessus biology and physiopathology.
Collapse
Affiliation(s)
- Ana Victoria Gutiérrez
- Centre National de la Recherche Scientifique, Institut de Recherche en Infectiologie de Montpellier, UMR 9004, Université de Montpellier, Montpellier, France.,CNRS, IRD 198, INSERM U1095, APHM, Institut Hospitalo-Universitaire Méditerranée Infection, UMR 7278, Aix-Marseille Université, Marseille, France
| | - Albertus Viljoen
- Centre National de la Recherche Scientifique, Institut de Recherche en Infectiologie de Montpellier, UMR 9004, Université de Montpellier, Montpellier, France
| | - Eric Ghigo
- CNRS, Campus Joseph Aiguier, Marseille, France
| | | | - Laurent Kremer
- Centre National de la Recherche Scientifique, Institut de Recherche en Infectiologie de Montpellier, UMR 9004, Université de Montpellier, Montpellier, France.,INSERM, IRIM, Montpellier, France
| |
Collapse
|
50
|
Nakayama T, Kawahara R, Kumeda Y, Yamamoto Y. Extended-spectrum β-lactamase-producing Escherichia coli contributes to the survival of cefotaxime-susceptible E. coli under high concentrations of cefotaxime by acquisition of increased AmpC expression. FEMS Microbiol Lett 2018; 365:4816729. [PMID: 29361027 DOI: 10.1093/femsle/fny009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 01/17/2018] [Indexed: 12/11/2022] Open
Abstract
Extended-spectrum β-lactamase-producing Escherichia coli (ESBL-E) are becoming increasingly widespread in Vietnam. Antibiotics are detected in many Vietnamese foods; however, the effect of ESBL-E and antibiotic consumption on intestinal bacteria has not been studied sufficiently. Here, we investigated the effect of oral administration of ESBL-E (TB19) and cefotaxime on luminescence-emitting cefotaxime-sensitive E. coli (X14). Mice were given water containing TB19 and then received three injections of 1.0 × 108 CFU of X14 harboring a luciferase gene. The mice were administered 100 μg of cefotaxime and luminescent bacteria were monitored over 24 h, following which luminescent bacteria were isolated from mouse feces. Luminescence continued to be detected in mice administered TB19 24 h after cefotaxime ingestion. Fecal analysis revealed two types of luminescent colonies: cefoxitin-resistant E. coli (X14-R) and Pseudomonas aeruginosa. Pulse-field gel electrophoresis confirmed that X14-R was a clonal strain of X14, suggesting that X14 survived using ESBLs originating from TB19 and acquired cefoxitin resistance due to cefotaxime consumption. Moreover, in vitro analysis of X14 indicated that expression of the ampC gene was upregulated by cefotaxime. Overall, ESBL-E and cefotaxime promoted the expansion of cefoxitin-resistant E. coli in the absence of plasmid-mediated gene transfer.
Collapse
Affiliation(s)
- Tatsuya Nakayama
- Division of Biomedical Food Research, National Institute of Health Science, Tokyo 158-8501, Japan.,Center for Global Initiatives, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ryuji Kawahara
- Department of Bacteriology, Osaka Institute of Public Health, Higashinari, Osaka 537-0025, Japan
| | - Yuko Kumeda
- Research Center for Microbial Control, Osaka Prefecture University, Osaka 565-8531, Japan
| | - Yoshimasa Yamamoto
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|