1
|
Yamaguchi N, Otsuna H, Eisenberg-Bord M, Ramakrishnan L. An Image Processing Tool for Automated Quantification of Bacterial Burdens in Zebrafish Larvae. Zebrafish 2025; 22:11-14. [PMID: 39718816 PMCID: PMC11971606 DOI: 10.1089/zeb.2024.0170] [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] [Indexed: 12/25/2024] Open
Abstract
Zebrafish larvae are used to model the pathogenesis of multiple bacteria. This transparent model offers the unique advantage of allowing quantification of fluorescent bacterial burdens (fluorescent pixel counts [FPC]) in vivo by facile microscopical methods, replacing enumeration of bacteria using time-intensive plating of lysates on bacteriological media. Accurate FPC measurements require laborious manual image processing to mark the outside borders of the animals so as to delineate the bacteria inside the animals from those in the culture medium that they are in. Here, we have developed an automated ImageJ/Fiji-based macro that accurately detects the outside borders of Mycobacterium marinum-infected larvae.
Collapse
Affiliation(s)
- Naoya Yamaguchi
- Department of Medicine, Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Hideo Otsuna
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Michal Eisenberg-Bord
- Department of Medicine, Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Lalita Ramakrishnan
- Department of Medicine, Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
- MRC Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
2
|
de Araujo MH, Muñoz Sánchez S, Simão TLBV, Nowik N, Antunes SS, Pinto SC, Sorze D, Boldrin F, Manganelli R, Correia Romeiro N, Lasunskaia EB, Verbeek FJ, Spaink HP, Muzitano MF. Exploring the Antimycobacterial Potential of Podocarpusflavone A from Kielmeyera membranacea: In Vitro and In Vivo Insights. Pharmaceuticals (Basel) 2024; 17:1560. [PMID: 39770402 PMCID: PMC11676425 DOI: 10.3390/ph17121560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Tuberculosis (TB) is one of the leading infectious causes of death worldwide, highlighting the importance of identifying new anti-TB agents. In previous research, our team identified antimycobacterial activity in Kielmeyera membranacea leaf extract; therefore, this study aims to conduct further exploration of its potential. Methods: Classical chromatography was applied for fractionation and spectrometric techniques were utilized for chemical characterization. For in vitro tests, samples were assessed against Mycobacterium tuberculosis and Mycobacterium marinum. The toxicity and efficacy of active samples were evaluated in vivo using different zebrafish models. Chemogenomics studies were applied to predict the isolated active compound's potential mode of action. Results: We performed fractionation of K. membranacea ethanolic extract (EE) and then its dichloromethane fraction (DCM), and the biflavonoid podocarpusflavone A (PCFA) was isolated and identified as a promising active compound. The EE and PCFA were found to be non-toxic to zebrafish larvae and were able to inhibit M. tuberculosis growth extracellularly. Additionally, PCFA demonstrated antimycobacterial activity within infected macrophages, especially when combined with isoniazid. In addition, the EE, DCM, and PCFA have shown the ability to inhibit M. marinum's growth during in vivo zebrafish larvae yolk infection. Notably, PCFA also effectively countered systemic infection established through the caudal vein, showing a similar inhibitory activity profile to rifampicin, both at 32 µM. A reduction in the transcriptional levels of pro-inflammatory cytokines confirmed the infection resolution. The protein tyrosine phosphatase B (PtpB) of M. tuberculosis, which inhibits the macrophage immune response, was predicted as a theoretical target of PCFA. This finding is in agreement with the higher activity observed for PCFA intracellularly and in vivo on zebrafish, compared with the direct action in M. tuberculosis. Conclusions: Here, we describe the discovery of PCFA as an intracellular inhibitor of M. tuberculosis and provide evidence of its in vivo efficacy and safety, encouraging its further development as a combination drug in novel therapeutic regimens for TB.
Collapse
Affiliation(s)
- Marlon Heggdorne de Araujo
- Laboratório de Produtos Bioativos (LPBio), Instituto de Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé 27930-560, RJ, Brazil; (M.H.d.A.); (S.S.A.); (S.C.P.)
- Department of Animal Sciences and Health, Institute of Biology (IBL), Leiden University, 2333 BE Leiden, The Netherlands; (S.M.S.); (H.P.S.)
| | - Salomé Muñoz Sánchez
- Department of Animal Sciences and Health, Institute of Biology (IBL), Leiden University, 2333 BE Leiden, The Netherlands; (S.M.S.); (H.P.S.)
| | - Thatiana Lopes Biá Ventura Simão
- Laboratório de Biologia do Reconhecer (LBR), Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes 28013-602, RJ, Brazil; (T.L.B.V.S.); (E.B.L.)
| | - Natalia Nowik
- Department of Animal Sciences and Health, Institute of Biology (IBL), Leiden University, 2333 BE Leiden, The Netherlands; (S.M.S.); (H.P.S.)
| | - Stella Schuenck Antunes
- Laboratório de Produtos Bioativos (LPBio), Instituto de Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé 27930-560, RJ, Brazil; (M.H.d.A.); (S.S.A.); (S.C.P.)
- Laboratório Integrado de Computação Científica (LICC), Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé 27930-560, RJ, Brazil;
| | - Shaft Corrêa Pinto
- Laboratório de Produtos Bioativos (LPBio), Instituto de Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé 27930-560, RJ, Brazil; (M.H.d.A.); (S.S.A.); (S.C.P.)
| | - Davide Sorze
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (D.S.); (F.B.); (R.M.)
| | - Francesca Boldrin
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (D.S.); (F.B.); (R.M.)
| | - Riccardo Manganelli
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (D.S.); (F.B.); (R.M.)
| | - Nelilma Correia Romeiro
- Laboratório Integrado de Computação Científica (LICC), Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé 27930-560, RJ, Brazil;
| | - Elena B. Lasunskaia
- Laboratório de Biologia do Reconhecer (LBR), Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes 28013-602, RJ, Brazil; (T.L.B.V.S.); (E.B.L.)
| | - Fons J. Verbeek
- Leiden Institute of Advanced Computer Science, Leiden University, 2333 CA Leiden, The Netherlands;
| | - Herman P. Spaink
- Department of Animal Sciences and Health, Institute of Biology (IBL), Leiden University, 2333 BE Leiden, The Netherlands; (S.M.S.); (H.P.S.)
| | - Michelle Frazão Muzitano
- Laboratório de Produtos Bioativos (LPBio), Instituto de Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé 27930-560, RJ, Brazil; (M.H.d.A.); (S.S.A.); (S.C.P.)
| |
Collapse
|
3
|
Antunes SS, Forn-Cuní G, Romeiro NC, Spaink HP, Verbeek FJ, Muzitano MF. Embryonic and larval zebrafish models for the discovery of new bioactive compounds against tuberculosis. Drug Discov Today 2024; 29:104163. [PMID: 39245344 DOI: 10.1016/j.drudis.2024.104163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/22/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
Tuberculosis (TB) is a world health challenge the treatment of which is impacted by the rise of drug-resistant strains. Thus, there is an urgent need for new antitubercular compounds and novel approaches to improve current TB therapy. The zebrafish animal model has become increasingly relevant as an experimental system. It has proven particularly useful during early development for aiding TB drug discovery, supporting both the discovery of new insights into mycobacterial pathogenesis and the evaluation of therapeutical toxicity and efficacy in vivo. In this review, we summarize the past two decades of zebrafish-Mycobacterium marinum research and discuss its contribution to the field of bioactive antituberculosis therapy development.
Collapse
Affiliation(s)
- Stella S Antunes
- Institute of Pharmaceutical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriel Forn-Cuní
- Institute of Biology Leiden, Leiden University, Leiden, the Netherlands
| | - Nelilma C Romeiro
- Institute of Pharmaceutical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Herman P Spaink
- Institute of Biology Leiden, Leiden University, Leiden, the Netherlands
| | - Fons J Verbeek
- Leiden Institute of Advanced Computer Science, Leiden University, Leiden, the Netherlands
| | - Michelle F Muzitano
- Institute of Pharmaceutical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
4
|
Yamaguchi N, Otsuna H, Eisenberg-Bord M, Ramakrishnan L. An Image Processing Tool for Automated Quantification of Bacterial Burdens in Zebrafish Larvae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608298. [PMID: 39229075 PMCID: PMC11370481 DOI: 10.1101/2024.08.16.608298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Zebrafish larvae are used to model the pathogenesis of multiple bacteria. This transparent model offers the unique advantage of allowing quantification of fluorescent bacterial burdens (fluorescent pixel counts: FPC) in vivo by facile microscopical methods, replacing enumeration of bacteria using time-intensive plating of lysates on bacteriological media. Accurate FPC measurements require laborious manual image processing to mark the outside borders of the animals so as to delineate the bacteria inside the animals from those in the culture medium that they are in. Here, we have developed an automated ImageJ/Fiji-based macro that accurately detect the outside borders of Mycobacterium marinum-infected larvae.
Collapse
Affiliation(s)
- Naoya Yamaguchi
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, CB2 0AW Cambridge, UK
- MRC Laboratory of Molecular Biology, CB2 0QH Cambridge, UK
- Equal contributions
| | - Hideo Otsuna
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
- Equal contributions
| | - Michal Eisenberg-Bord
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, CB2 0AW Cambridge, UK
- MRC Laboratory of Molecular Biology, CB2 0QH Cambridge, UK
| | - Lalita Ramakrishnan
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, CB2 0AW Cambridge, UK
- MRC Laboratory of Molecular Biology, CB2 0QH Cambridge, UK
| |
Collapse
|
5
|
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
|
6
|
Xia J, Wang H, Zhong Z, Jiang J. Inhibition of PIKfyve Leads to Lysosomal Disorders via Dysregulation of mTOR Signaling. Cells 2024; 13:953. [PMID: 38891085 PMCID: PMC11171791 DOI: 10.3390/cells13110953] [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: 04/24/2024] [Revised: 05/23/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
PIKfyve is an endosomal lipid kinase that synthesizes phosphatidylinositol 3,5-biphosphate from phosphatidylinositol 3-phsphate. Inhibition of PIKfyve activity leads to lysosomal enlargement and cytoplasmic vacuolation, attributed to impaired lysosomal fission processes and homeostasis. However, the precise molecular mechanisms underlying these effects remain a topic of debate. In this study, we present findings from PIKfyve-deficient zebrafish embryos, revealing enlarged macrophages with giant vacuoles reminiscent of lysosomal storage disorders. Treatment with mTOR inhibitors or effective knockout of mTOR partially reverses these abnormalities and extend the lifespan of mutant larvae. Further in vivo and in vitro mechanistic investigations provide evidence that PIKfyve activity is essential for mTOR shutdown during early zebrafish development and in cells cultured under serum-deprived conditions. These findings underscore the critical role of PIKfyve activity in regulating mTOR signaling and suggest potential therapeutic applications of PIKfyve inhibitors for the treatment of lysosomal storage disorders.
Collapse
Affiliation(s)
- Jianhong Xia
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; (J.X.); (H.W.)
| | - Haiyun Wang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; (J.X.); (H.W.)
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Zhihang Zhong
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou 510642, China;
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Jun Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou 510642, China;
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
7
|
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
|
8
|
Fröhlich E. Animals in Respiratory Research. Int J Mol Sci 2024; 25:2903. [PMID: 38474149 DOI: 10.3390/ijms25052903] [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: 01/17/2024] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
The respiratory barrier, a thin epithelial barrier that separates the interior of the human body from the environment, is easily damaged by toxicants, and chronic respiratory diseases are common. It also allows the permeation of drugs for topical treatment. Animal experimentation is used to train medical technicians, evaluate toxicants, and develop inhaled formulations. Species differences in the architecture of the respiratory tract explain why some species are better at predicting human toxicity than others. Some species are useful as disease models. This review describes the anatomical differences between the human and mammalian lungs and lists the characteristics of currently used mammalian models for the most relevant chronic respiratory diseases (asthma, chronic obstructive pulmonary disease, cystic fibrosis, pulmonary hypertension, pulmonary fibrosis, and tuberculosis). The generation of animal models is not easy because they do not develop these diseases spontaneously. Mouse models are common, but other species are more appropriate for some diseases. Zebrafish and fruit flies can help study immunological aspects. It is expected that combinations of in silico, in vitro, and in vivo (mammalian and invertebrate) models will be used in the future for drug development.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, 8010 Graz, Austria
- Research Center Pharmaceutical Engineering GmbH, 8010 Graz, Austria
| |
Collapse
|
9
|
Wen D, Meng C, Feng Y, Shen L, Liu Y, Sun W, Chen G, Wu C. Syringaldehyde Exhibits Antibacterial and Antioxidant Activities against Mycobacterium marinum Infection. Microorganisms 2024; 12:348. [PMID: 38399751 PMCID: PMC10893232 DOI: 10.3390/microorganisms12020348] [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: 01/09/2024] [Revised: 01/27/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Tuberculosis (TB) is caused by infection with Mycobacterium tuberculosis (Mtb), which has a unique resistance to many antimicrobial agents. TB has emerged as a significant worldwide health issue because of the rise of multidrug-resistant strains causing drug-resistant TB (DR-TB). As a result, the development of new drugs or effective strategies is crucial for patients with TB. Mycobacterium marinum (Mm) and Mtb are both species of mycobacteria. In zebrafish, Mm proliferates and forms chronic granulomatous infections, which are similar to Mtb infections in lung tissue. Syringaldehyde (SA) is a member of the phenolic aldehyde family found in various plants. Here, we investigated its antioxidative and antibacterial properties in Mm-infected cells and zebrafish. Our results demonstrated that SA inhibits Mm-infected pulmonary epithelial cells and inhibits the proliferation of Mm in Mm-infected zebrafish, suggesting that SA provides an antibacterial effect during Mm infection. Further study demonstrated that supplementation with SA inhibits the production of malondialdehyde (MDA) and reactive oxygen species (ROS) and increases the levels of reduced glutathione (GSH) in Mm-infection-induced macrophages. SA inhibits the levels of MDA in Mm-infected zebrafish, suggesting that SA exerts antioxidative effects in vivo. Additionally, we found that SA promotes the expression of NRF2/HO-1/NQO-1 and the activation of the AMPK-α1/AKT/GSK-3β signaling pathway. In summary, our data demonstrated that SA exerts antioxidative and antibacterial effects during Mm infection both in vivo and in vitro and that the antioxidative effects of SA may be due to the regulation of NRF2/HO-1/NQO-1 and the AMPK-α1/AKT/GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Da Wen
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Chaoqun Meng
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Yazhi Feng
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Lin Shen
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Yiyao Liu
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Wei Sun
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Guangxin Chen
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory for Prevention and Treatment of Major Infectious Diseases, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
10
|
Bloom BR. A half-century of research on tuberculosis: Successes and challenges. J Exp Med 2023; 220:e20230859. [PMID: 37552470 PMCID: PMC10407785 DOI: 10.1084/jem.20230859] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/09/2023] Open
Abstract
Great progress has been made over the past half-century, but TB remains a formidable global health problem, particularly in low- and middle-income countries. Understanding the mechanisms of pathogenesis and necessary and sufficient conditions for protection are critical. The need for inexpensive and sensitive point-of-care diagnostic tests for earlier detection of infection and disease, shorter and less-toxic drug regimens for drug-sensitive and -resistant TB, and a more effective vaccine than BCG is immense. New and better tools, greater support for international research, collaborations, and training will be required to dramatically reduce the burden of this devastating disease which still kills 1.6 million people annually.
Collapse
Affiliation(s)
- Barry R. Bloom
- Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
11
|
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
|
12
|
Dal NJK, Schäfer G, Thompson AM, Schmitt S, Redinger N, Alonso-Rodriguez N, Johann K, Ojong J, Wohlmann J, Best A, Koynov K, Zentel R, Schaible UE, Griffiths G, Barz M, Fenaroli F. Π-Π interactions stabilize PeptoMicelle-based formulations of Pretomanid derivatives leading to promising therapy against tuberculosis in zebrafish and mouse models. J Control Release 2023; 354:851-868. [PMID: 36681282 DOI: 10.1016/j.jconrel.2023.01.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/15/2022] [Accepted: 01/14/2023] [Indexed: 01/23/2023]
Abstract
Tuberculosis is the deadliest bacterial disease globally, threatening the lives of millions every year. New antibiotic therapies that can shorten the duration of treatment, improve cure rates, and impede the development of drug resistance are desperately needed. Here, we used polymeric micelles to encapsulate four second-generation derivatives of the antitubercular drug pretomanid that had previously displayed much better in vivo activity against Mycobacterium tuberculosis than pretomanid itself. Because these compounds were relatively hydrophobic and had limited bioavailability, we expected that their micellar formulations would overcome these limitations, reduce toxicities, and improve therapeutic outcomes. The polymeric micelles were based on polypept(o)ides (PeptoMicelles) and were stabilized in their hydrophobic core by π-π interactions, allowing the efficient encapsulation of aromatic pretomanid derivatives. The stability of these π-π-stabilized PeptoMicelles was demonstrated in water, blood plasma, and lung surfactant by fluorescence cross-correlation spectroscopy and was further supported by prolonged circulation times of several days in the vasculature of zebrafish larvae. The most efficacious PeptoMicelle formulation tested in the zebrafish larvae infection model almost completely eradicated the bacteria at non-toxic doses. This lead formulation was further assessed against Mycobacterium tuberculosis in the susceptible C3HeB/FeJ mouse model, which develops human-like necrotic granulomas. Following intravenous administration, the drug-loaded PeptoMicelles significantly reduced bacterial burden and inflammatory responses in the lungs and spleens of infected mice.
Collapse
Affiliation(s)
- Nils-Jørgen K Dal
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Gabriela Schäfer
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany; Leiden Academic Center for Drug Research (LACDR), Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC, Leiden, the Netherlands
| | - Andrew M Thompson
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Sascha Schmitt
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Natalja Redinger
- Forschungszentrum Borstel, Leibniz Lungenzentrum, Program Area Infections, Div. Cellular Microbiology; University of Lübeck, Immunochemistry and Biochemical Microbiology, & German Center for Infection Research, partner site Hamburg-Lübeck - Borstel - Riems, 23845 Borstel, Germany
| | | | - Kerstin Johann
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Jessica Ojong
- Forschungszentrum Borstel, Leibniz Lungenzentrum, Program Area Infections, Div. Cellular Microbiology; University of Lübeck, Immunochemistry and Biochemical Microbiology, & German Center for Infection Research, partner site Hamburg-Lübeck - Borstel - Riems, 23845 Borstel, Germany
| | - Jens Wohlmann
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Andreas Best
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Kaloian Koynov
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Rudolf Zentel
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Ulrich E Schaible
- Forschungszentrum Borstel, Leibniz Lungenzentrum, Program Area Infections, Div. Cellular Microbiology; University of Lübeck, Immunochemistry and Biochemical Microbiology, & German Center for Infection Research, partner site Hamburg-Lübeck - Borstel - Riems, 23845 Borstel, Germany
| | - Gareth Griffiths
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Matthias Barz
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany; Leiden Academic Center for Drug Research (LACDR), Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC, Leiden, the Netherlands.
| | - Federico Fenaroli
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway; Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4021 Stavanger, Norway.
| |
Collapse
|
13
|
Dam S, Tangara S, Hamela C, Hattabi T, Faïon L, Carre P, Antoine R, Herledan A, Leroux F, Piveteau C, Eveque M, Flipo M, Deprez B, Kremer L, Willand N, Villemagne B, Hartkoorn RC. Tricyclic SpiroLactams Kill Mycobacteria In Vitro and In Vivo by Inhibiting Type II NADH Dehydrogenases. J Med Chem 2022; 65:16651-16664. [PMID: 36473699 PMCID: PMC9791652 DOI: 10.1021/acs.jmedchem.2c01493] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is critical that novel classes of antituberculosis drugs are developed to combat the increasing burden of infections by multidrug-resistant strains. To identify such a novel class of antibiotics, a chemical library of unique 3-D bioinspired molecules was explored revealing a promising, mycobacterium specific Tricyclic SpiroLactam (TriSLa) hit. Chemical optimization of the TriSLa scaffold delivered potent analogues with nanomolar activity against replicating and nonreplicating Mycobacterium tuberculosis. Characterization of isolated TriSLa-resistant mutants, and biochemical studies, found TriSLas to act as allosteric inhibitors of type II NADH dehydrogenases (Ndh-2 of the electron transport chain), resulting in an increase in bacterial NADH/NAD+ ratios and decreased ATP levels. TriSLas are chemically distinct from other inhibitors of Ndh-2 but share a dependence for fatty acids for activity. Finally, in vivo proof-of-concept studies showed TriSLas to protect zebrafish larvae from Mycobacterium marinum infection, suggesting a vulnerability of Ndh-2 inhibition in mycobacterial infections.
Collapse
Affiliation(s)
- Sushovan Dam
- Univ.
Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR
9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Salia Tangara
- Univ.
Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules
for Living Systems, F-59000 Lille, France
| | - Claire Hamela
- Centre
National de la Recherche Scientifique, Institut de Recherche en Infectiologie
de Montpellier, UMR 9004, Université
de Montpellier, 34293 Montpellier, France
| | - Theo Hattabi
- Univ.
Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules
for Living Systems, F-59000 Lille, France
| | - Léo Faïon
- Univ.
Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules
for Living Systems, F-59000 Lille, France
| | - Paul Carre
- Univ.
Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR
9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Rudy Antoine
- Univ.
Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR
9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Adrien Herledan
- Univ.
Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules
for Living Systems, F-59000 Lille, France
| | - Florence Leroux
- Univ.
Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules
for Living Systems, F-59000 Lille, France
| | - Catherine Piveteau
- Univ.
Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules
for Living Systems, F-59000 Lille, France
| | - Maxime Eveque
- Univ.
Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules
for Living Systems, F-59000 Lille, France
| | - Marion Flipo
- Univ.
Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules
for Living Systems, F-59000 Lille, France
| | - Benoit Deprez
- Univ.
Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules
for Living Systems, F-59000 Lille, France
| | - Laurent Kremer
- Centre
National de la Recherche Scientifique, Institut de Recherche en Infectiologie
de Montpellier, UMR 9004, Université
de Montpellier, 34293 Montpellier, France,INSERM, IRIM, 34293 Montpellier, France
| | - Nicolas Willand
- Univ.
Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules
for Living Systems, F-59000 Lille, France,
| | - Baptiste Villemagne
- Univ.
Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules
for Living Systems, F-59000 Lille, France,
| | - Ruben C. Hartkoorn
- Univ.
Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR
9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France,
| |
Collapse
|
14
|
He S, Fan H, Sun B, Yang M, Liu H, Yang J, Liu J, Luo S, Chen Z, Zhou J, Xia L, Zhang S, Yan B. Tibetan medicine salidroside improves host anti-mycobacterial response by boosting inflammatory cytokine production in zebrafish. Front Pharmacol 2022; 13:936295. [PMID: 36120339 PMCID: PMC9470765 DOI: 10.3389/fphar.2022.936295] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/29/2022] [Indexed: 12/02/2022] Open
Abstract
The treatment for tuberculosis (TB), especially multidrug-resistant TB (MDR-TB), has a prolonged cycle which can last up to a year. This is partially due to the lack of effective therapies. The development of novel anti-TB drugs from the perspective of host immune regulation can provide an important supplement for conventional treatment strategies. Salidroside (SAL), a bioactive component from the Tibetan medicine Rhodiola rosea, has been used in the treatment of TB, although its mechanism remains unclear. Here, the bacteriostatic effect of SAL in vivo was first demonstrated using a zebrafish–M. marinum infection model. To further investigate the underlying mechanism, we then examined the impact of SAL on immune cell recruitment during wound and infection. Increased macrophage and neutrophil infiltrations were found both in the vicinity of the wound and infection sites after SAL treatment compared with control, which might be due to the elevated chemokine expression levels after SAL treatment. SAL treatment alone was also demonstrated to improve the survival of infected zebrafish larvae, an effect that was amplified when combining SAL treatment with isoniazid or rifampicin. Interestingly, the reduced bacterial burden and improved survival rate under SAL treatment were compromised in tnfα-deficient embryos which suggests a requirement of Tnfα signaling on the anti-mycobacterial effects of SAL. In summary, this study provides not only the cellular and molecular mechanisms for the host anti-mycobacterial effects of the Tibetan medicine SAL but also proof of concept that combined application of SAL with traditional first-line anti-TB drugs could be a novel strategy to improve treatment efficacy.
Collapse
Affiliation(s)
- Shumei He
- Key Laboratory of Molecular Mechanistic and Interventional Research of Plateau Diseases in Tibet Autonomous Region, Key Laboratory of High Altitude Hypoxia Environment and Life Health, Joint Central Laboratory for Active Components and Pharmacological Mechanism of Tibetan Medicine, School of Medicine, Xizang Minzu University, Xianyang, China
- *Correspondence: Shumei He, ; Shulin Zhang, ; Bo Yan,
| | - Hongyan Fan
- Key Laboratory of Molecular Mechanistic and Interventional Research of Plateau Diseases in Tibet Autonomous Region, Key Laboratory of High Altitude Hypoxia Environment and Life Health, Joint Central Laboratory for Active Components and Pharmacological Mechanism of Tibetan Medicine, School of Medicine, Xizang Minzu University, Xianyang, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Bin Sun
- Department of Stomatology, The First Affiliated Hospital of Shihezi University Medical College, Shihezi, China
| | - Meipan Yang
- Key Laboratory of Molecular Mechanistic and Interventional Research of Plateau Diseases in Tibet Autonomous Region, Key Laboratory of High Altitude Hypoxia Environment and Life Health, Joint Central Laboratory for Active Components and Pharmacological Mechanism of Tibetan Medicine, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Hongxu Liu
- Key Laboratory of Molecular Mechanistic and Interventional Research of Plateau Diseases in Tibet Autonomous Region, Key Laboratory of High Altitude Hypoxia Environment and Life Health, Joint Central Laboratory for Active Components and Pharmacological Mechanism of Tibetan Medicine, School of Medicine, Xizang Minzu University, Xianyang, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jianwei Yang
- Key Laboratory of Molecular Mechanistic and Interventional Research of Plateau Diseases in Tibet Autonomous Region, Key Laboratory of High Altitude Hypoxia Environment and Life Health, Joint Central Laboratory for Active Components and Pharmacological Mechanism of Tibetan Medicine, School of Medicine, Xizang Minzu University, Xianyang, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jianxin Liu
- Key Laboratory of Molecular Mechanistic and Interventional Research of Plateau Diseases in Tibet Autonomous Region, Key Laboratory of High Altitude Hypoxia Environment and Life Health, Joint Central Laboratory for Active Components and Pharmacological Mechanism of Tibetan Medicine, School of Medicine, Xizang Minzu University, Xianyang, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Sizhu Luo
- Key Laboratory of Molecular Mechanistic and Interventional Research of Plateau Diseases in Tibet Autonomous Region, Key Laboratory of High Altitude Hypoxia Environment and Life Health, Joint Central Laboratory for Active Components and Pharmacological Mechanism of Tibetan Medicine, School of Medicine, Xizang Minzu University, Xianyang, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zihan Chen
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Medical College, China Three Gorges University, Yichang, China
| | - Jing Zhou
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Medical College, China Three Gorges University, Yichang, China
| | - Lu Xia
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Shulin Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Shumei He, ; Shulin Zhang, ; Bo Yan,
| | - Bo Yan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- *Correspondence: Shumei He, ; Shulin Zhang, ; Bo Yan,
| |
Collapse
|
15
|
Amaning Danquah C, Minkah PAB, Osei Duah Junior I, Amankwah KB, Somuah SO. Antimicrobial Compounds from Microorganisms. Antibiotics (Basel) 2022; 11:285. [PMID: 35326749 PMCID: PMC8944786 DOI: 10.3390/antibiotics11030285] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/27/2022] [Accepted: 02/07/2022] [Indexed: 02/06/2023] Open
Abstract
Antimicrobial resistance is an exigent public health concern owing to the emergence of novel strains of human resistant pathogens and the concurrent rise in multi-drug resistance. An influx of new antimicrobials is urgently required to improve the treatment outcomes of infectious diseases and save lives. Plant metabolites and bioactive compounds from chemical synthesis have found their efficacy to be dwindling, despite some of them being developed as drugs and used to treat human infections for several decades. Microorganisms are considered untapped reservoirs for promising biomolecules with varying structural and functional antimicrobial activity. The advent of cost-effective and convenient model organisms, state-of-the-art molecular biology, omics technology, and machine learning has enhanced the bioprospecting of novel antimicrobial drugs and the identification of new drug targets. This review summarizes antimicrobial compounds isolated from microorganisms and reports on the modern tools and strategies for exploiting promising antimicrobial drug candidates. The investigation identified a plethora of novel compounds from microbial sources with excellent antimicrobial activity against disease-causing human pathogens. Researchers could maximize the use of novel model systems and advanced biomolecular and computational tools in exploiting lead antimicrobials, consequently ameliorating antimicrobial resistance.
Collapse
Affiliation(s)
- Cynthia Amaning Danquah
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, PMB, Kumasi, Ghana;
| | - Prince Amankwah Baffour Minkah
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, PMB, Kumasi, Ghana;
- Global Health and Infectious Disease Research Group, Kumasi Centre for Collaborative Research in Tropical Medicine, College of Health Sciences, Kwame Nkrumah University of Science and Technology, PMB, Kumasi, Ghana
| | - Isaiah Osei Duah Junior
- Department of Optometry and Visual Science, College of Science, Kwame Nkrumah University of Science and Technology, PMB, Kumasi, Ghana;
| | - Kofi Bonsu Amankwah
- Department of Biomedical Sciences, University of Cape Coast, PMB, Cape Coast, Ghana;
| | - Samuel Owusu Somuah
- Department of Pharmacy Practice, School of Pharmacy, University of Health and Allied Sciences, PMB, Ho, Ghana;
| |
Collapse
|
16
|
Knudsen Dal NJ, Speth M, Johann K, Barz M, Beauvineau C, Wohlmann J, Fenaroli F, Gicquel B, Griffiths G, Alonso-Rodriguez N. The zebrafish embryo as an in vivo model for screening nanoparticle-formulated lipophilic anti-tuberculosis compounds. Dis Model Mech 2022; 15:dmm049147. [PMID: 34842273 PMCID: PMC8807572 DOI: 10.1242/dmm.049147] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 11/16/2021] [Indexed: 11/20/2022] Open
Abstract
With the increasing emergence of drug-resistant Mycobacterium tuberculosis strains, new and effective antibiotics against tuberculosis (TB) are urgently needed. However, the high frequency of poorly water-soluble compounds among hits in high-throughput drug screening campaigns is a major obstacle in drug discovery. Moreover, in vivo testing using conventional animal TB models, such as mice, is time consuming and costly, and represents a major bottleneck in lead compound discovery and development. Here, we report the use of the zebrafish embryo TB model for evaluating the in vivo toxicity and efficacy of five poorly water-soluble nitronaphthofuran derivatives, which were recently identified as possessing anti-TB activity in vitro. To aid solubilization, compounds were formulated in biocompatible polymeric micelles (PMs). Three of the five PM-formulated nitronaphthofuran derivatives showed low toxicity in vivo, significantly reduced bacterial burden and improved survival in infected zebrafish embryos. We propose the zebrafish embryo TB-model as a quick and sensitive tool for evaluating the in vivo toxicity and efficacy of new anti-TB compounds during early stages of drug development. Thus, this model is well suited for pinpointing promising compounds for further development.
Collapse
Affiliation(s)
- Nils-Jørgen Knudsen Dal
- Department Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0371 Oslo, Norway
| | - Martin Speth
- Department Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0371 Oslo, Norway
| | - Kerstin Johann
- Department of Chemistry, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Matthias Barz
- Department of Chemistry, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- Division of BioTherapeutics, Leiden Academic Center for Drug Research (LACDR), Leiden University, 2333 Leiden, The Netherlands
| | - Claire Beauvineau
- Chemical Library Institut Curie/CNRS, CNRS UMR9187, INSERM U1196 and CNRS UMR3666, INSERM U1193, Université Paris-Saclay, F-91405 Orsay, France
| | - Jens Wohlmann
- Department Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0371 Oslo, Norway
| | - Federico Fenaroli
- Department Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0371 Oslo, Norway
| | - Brigitte Gicquel
- Unité de Génétique Mycobactérienne, Dep Génomes and Génétique, Institute Pasteur, 75015 Paris, France
- Department of Tuberculosis Control and Prevention, Shenzhen Nanshan Center for Chronic Disease Control, 518054 Shenzhen, China
| | - Gareth Griffiths
- Department Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0371 Oslo, Norway
| | - Noelia Alonso-Rodriguez
- Department Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0371 Oslo, Norway
| |
Collapse
|
17
|
Lian B, Li Y, Yang Q, Xie L, Zhang Q, Liu Y, Zhao X, Li S. Phloretin loaded porous starch (Ph-PS): Preparation, characterization, in vitro release and protective effect against oxidative stress in vivo zebrafish model. Int J Biol Macromol 2021; 193:2047-2053. [PMID: 34774597 DOI: 10.1016/j.ijbiomac.2021.11.036] [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: 08/09/2021] [Revised: 10/17/2021] [Accepted: 11/04/2021] [Indexed: 11/15/2022]
Abstract
Phloretin loaded porous starch (Ph-PS) were prepared for its application in food. The effects of Ph-PS in vitro release and its ability against AAPH-induced oxidative stress in vivo zebrafish model were investigated. Ph-PS was prepared by absorption method, the physical and chemical characterization showed that PS decreased the crystallinity of Ph obviously. Ph-PS exhibited higher release amount and faster release rate of Ph compared to free Ph in vitro release study. What's more, the effect of Ph-PS reduced ROS generation and lipid peroxidation was better than that of free Ph in zebrafish model. These findings suggest Ph-PS is a new and simple strategy to improve dissolution rate and antioxidant ability of Ph.
Collapse
Affiliation(s)
- Bolin Lian
- School of Life Sciences, Nantong University, Nantong 226019, Jiangsu, China
| | - Yuanyuan Li
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, Heilongjiang, China; Key Laboratory of Bio-Based Material Science and Technology of Ministry of Education, Northeast Forestry University, Harbin 150040, Heilongjiang, China
| | - Qilei Yang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, Heilongjiang, China
| | - Lanlan Xie
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, Heilongjiang, China
| | - Qian Zhang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, Heilongjiang, China
| | - Yanjie Liu
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, Heilongjiang, China
| | - Xiuhua Zhao
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Ecological Utilization of Forestry-Based Active Substances, China.
| | - Shujun Li
- Key Laboratory of Bio-Based Material Science and Technology of Ministry of Education, Northeast Forestry University, Harbin 150040, Heilongjiang, China.
| |
Collapse
|
18
|
Habjan E, Ho VQT, Gallant J, Van Stempvoort G, Jim KK, Kuijl C, Geerke DP, Bitter W, Speer A. Anti-tuberculosis Compound Screen using a Zebrafish Infection Model identifies an Aspartyl-tRNA Synthetase Inhibitor. Dis Model Mech 2021; 14:273850. [PMID: 34643222 PMCID: PMC8713996 DOI: 10.1242/dmm.049145] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 10/03/2021] [Indexed: 11/20/2022] Open
Abstract
Finding new anti-tuberculosis compounds with convincing in vivo activity is an ongoing global challenge to fight the emergence of multidrug-resistant Mycobacterium tuberculosis isolates. In this study, we exploited the medium-throughput capabilities of the zebrafish embryo infection model with Mycobacterium marinum as a surrogate for M. tuberculosis. Using a representative set of clinically established drugs, we demonstrate that this model could be predictive and selective for antibiotics that can be administered orally. We further used the zebrafish infection model to screen 240 compounds from an anti-tuberculosis hit library for their in vivo activity and identified 14 highly active compounds. One of the most active compounds was the tetracyclic compound TBA161, which was studied in more detail. Analysis of resistant mutants revealed point mutations in aspS (rv2572c), encoding an aspartyl-tRNA synthetase. The target was genetically confirmed, and molecular docking studies propose the possible binding of TBA161 in a pocket adjacent to the catalytic site. This study shows that the zebrafish infection model is suitable for rapidly identifying promising scaffolds with in vivo activity. Summary: Exploitation of the medium-throughput capabilities of a zebrafish embryo infection model of tuberculosis to screen compounds for their in vivo activity, one of which was characterized as an aspartyl-tRNA synthetase inhibitor.
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.,Section Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Vien Q T Ho
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - James Gallant
- Section Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Gunny Van Stempvoort
- Section Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Kin Ki Jim
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Coen Kuijl
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Daan P Geerke
- Department of Molecular Toxicology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ 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, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, 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
| |
Collapse
|
19
|
Zhou W, Yang B, Zou Y, Rahman K, Cao X, Lei Y, Lai R, Fu ZF, Chen X, Cao G. Screening of Compounds for Anti-tuberculosis Activity, and in vitro and in vivo Evaluation of Potential Candidates. Front Microbiol 2021; 12:658637. [PMID: 34276592 PMCID: PMC8278749 DOI: 10.3389/fmicb.2021.658637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB) is a debilitating infectious disease responsible for more than one million deaths per year. The emergence of drug-resistant TB poses an urgent need for the development of new anti-TB drugs. In this study, we screened a library of over 4,000 small molecules and found that orbifloxacin and the peptide AK15 possess significant bactericidal activity against Mycobacterium tuberculosis (Mtb) in vitro. Orbifloxacin also showed an effective ability on the clearance of intracellular Mtb and protect mice from a strong inflammatory response but not AK15. Moreover, we identified 17 nucleotide mutations responsible for orbifloxacin resistance by whole-genome sequencing. A critical point mutation (D94G) of the DNA gyrase (gyrA) gene was found to be the key role of resistance to orbifloxacin. The computational docking revealed that GyrA D94G point mutation can disrupt the orbifloxacin–protein gyrase interactions mediated by magnesium ion bridge. Overall, this study indicated the potential ability of orbifloxacin as an anti-tuberculosis drug, which can be used either alone or in combination with first-line antibiotics to achieve more effective therapy on TB.
Collapse
Affiliation(s)
- Wei Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Bing Yang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yanyan Zou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Khaista Rahman
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaojian Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yingying Lei
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Zhen F Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xi Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Gang Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Bio-Medical Center, Huazhong Agricultural University, Wuhan, China.,Cooperative Innovation Center for Sustainable Pig Production (CICSPPS), Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
20
|
Comparison of gene expression responses of zebrafish larvae to Vibrio parahaemolyticus infection by static immersion and caudal vein microinjection. AQUACULTURE AND FISHERIES 2021. [DOI: 10.1016/j.aaf.2019.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
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
|
22
|
Analysis tools to quantify dissemination of pathology in zebrafish larvae. Sci Rep 2020; 10:3149. [PMID: 32081863 PMCID: PMC7035342 DOI: 10.1038/s41598-020-59932-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 02/03/2020] [Indexed: 12/16/2022] Open
Abstract
We describe new open source software called QuantiFish for rapid quantitation of fluorescent foci in zebrafish larvae, to support infection research in this animal model. QuantiFish extends the conventional measurements of bacterial load and number of bacterial foci to include measures for dissemination of infection. These are represented by the proportions of bacteria between foci and their spatial distribution. We showcase these measures by comparison of intravenous and hindbrain routes of Mycobacterium marinum infection, which are indistinguishable by measurement of bacterial load and not consistently differentiated by the number of bacterial foci. The intravenous route showed dose dependent dissemination of infection, reflected by increased spatial dispersion of bacteria and lower proportions of bacteria distributed across many foci. In contrast, hindbrain infection resulted in localised disease, limited to a smaller area and higher proportions of bacteria distributed across fewer foci. The application of QuantiFish may extend beyond models of infection, to study other pathologies such as metastatic cancer.
Collapse
|
23
|
Parish T. In vitro drug discovery models for Mycobacterium tuberculosis relevant for host infection. Expert Opin Drug Discov 2020; 15:349-358. [PMID: 31899974 DOI: 10.1080/17460441.2020.1707801] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Introduction: Tuberculosis is the leading cause of death from infectious disease. Current drug therapy requires a combination of antibiotics taken over >6 months. An urgent need for new agents that can shorten therapy is required. In order to develop new drugs, simple in vitro assays are required that can identify efficacious compounds rapidly and predict in vivo activity in the human.Areas covered: This review focusses on the most relevant in vitro assays that can be utilized in a drug discovery program and which mimic different aspects of infection or disease. The focus is largely on assays used to test >1000s of compounds reliably and robustly. However, some assays used for 10s to 100s of compounds are included where the utility outweighs the low capacity. Literature searches for high throughput screening, models and in vitro assays were undertaken.Expert opinion: Drug discovery and development in tuberculosis is extremely challenging due to the requirement for predicting drug efficacy in a disease with complex pathology in which bacteria exist in heterogeneous states in inaccesible locations. A combination of assays can be used to determine profiles against replicating, non-replicating, intracellular and tolerant bacteria.
Collapse
Affiliation(s)
- Tanya Parish
- Seattle Children's Research Institute, Center for Global Infectious Disease Research, Seattle, WA, USA
| |
Collapse
|
24
|
Animal Models of Tuberculosis Vaccine Research: An Important Component in the Fight against Tuberculosis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4263079. [PMID: 32025519 PMCID: PMC6984742 DOI: 10.1155/2020/4263079] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/25/2019] [Accepted: 08/20/2019] [Indexed: 12/23/2022]
Abstract
Tuberculosis (TB), an infectious disease caused by Mycobacterium tuberculosis, is one of the top ten infectious diseases worldwide, and is the leading cause of morbidity from a single infectious agent. M. tuberculosis can cause infection in several species of animals in addition to humans as the natural hosts. Although animal models of TB disease cannot completely simulate the occurrence and development of human TB, they play an important role in studying the pathogenesis, immune responses, and pathological changes as well as for vaccine research. This review summarizes the commonly employed animal models, including mouse, guinea pig, rabbit, rat, goat, cattle, and nonhuman primates, and their characteristics as used in TB vaccine research, and provides a basis for selecting appropriate animal models according to specific research needs. Furthermore, some of the newest animal models used for TB vaccine research (such as humanized animal models, zebrafish, Drosophila, and amoeba) are introduced, and their characteristics and research progress are discussed.
Collapse
|
25
|
Cheng T, Kam JY, Johansen MD, Oehlers SH. High content analysis of granuloma histology and neutrophilic inflammation in adult zebrafish infected with Mycobacterium marinum. Micron 2019; 129:102782. [PMID: 31775097 DOI: 10.1016/j.micron.2019.102782] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022]
Abstract
Infection of zebrafish with natural pathogen Mycobacterium marinum is a useful surrogate for studying the human granulomatous inflammatory response to infection by Mycobacterium tuberculosis. The adaptive immune system of the adult stage zebrafish offers an advance on the commonly used embryo infection model as adult zebrafish form granulomas with striking similarities to human-M. tuberculosis granulomas. Here, we present workflows to perform high content analyses of granulomas in adult zebrafish infected with M. marinum by cryosectioning to take advantage of strong endogenous transgenic fluorescence adapted from common zebrafish embryo infection tools. Specific guides to classifying granuloma necrosis and organisation, quantifying bacterial burden and leukocyte infiltration of granulomas, visualizing foam cell formation, analysing extracellular matrix remodelling and granuloma fibrosis are also provided. We use these methods to characterize neutrophil recruitment to M. marinum granulomas across time and find an inverse relation to granuloma necrosis suggesting granuloma necrosis is not a marker of immunopathology in the natural infection system of the adult zebrafish-M. marinum pairing. The methods can be easily translated to studying the zebrafish adaptive immune response to other chronic and granuloma-forming pathogens.
Collapse
Affiliation(s)
- Tina Cheng
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Julia Y Kam
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Matt D Johansen
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, 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, Discipline of Infectious Diseases & Immunology and Marie Bashir Institute, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
26
|
Monitoring Tuberculosis Drug Activity in Live Animals by Near-Infrared Fluorescence Imaging. Antimicrob Agents Chemother 2019:AAC.01280-19. [PMID: 31527027 DOI: 10.1128/aac.01280-19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Worldwide, tuberculosis (TB) is the leading cause of death due to infection with a single pathogenic agent, Mycobacterium tuberculosis In the absence of an effective vaccine, new, more powerful antibiotics are required to halt the growing spread of multidrug-resistant strains and to shorten the duration of TB treatment. However, assessing drug efficacy at the preclinical stage remains a long and fastidious procedure that delays progression of drugs down the pipeline and towards the clinic. In this investigation, we report the construction, optimization and characterization of genetically engineered near-infrared (NIR) fluorescent reporter strains of the pathogens Mycobacterium marinum and Mycobacterium tuberculosis that enable direct visualization of bacteria in infected zebrafish and mice, respectively. Fluorescence could be measured precisely in infected immunodeficient mice, while its intensity appeared to be below the limit of detection in immunocompetent mice, probably because of the lower bacterial load obtained in these animals. Furthermore, we show that the fluorescence level accurately reflects the bacterial load, as determined by colony forming unit (CFU) enumeration, thus enabling the efficacy of antibiotic treatment to be assessed in live animals in real time. The NIR fluorescent imaging system disclosed here is a valuable resource for TB research and can serve to accelerate drug development.
Collapse
|
27
|
Hsu AY, Wang D, Liu S, Lu J, Syahirah R, Bennin DA, Huttenlocher A, Umulis DM, Wan J, Deng Q. Phenotypical microRNA screen reveals a noncanonical role of CDK2 in regulating neutrophil migration. Proc Natl Acad Sci U S A 2019; 116:18561-18570. [PMID: 31451657 PMCID: PMC6744913 DOI: 10.1073/pnas.1905221116] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neutrophil migration is essential for inflammatory responses to kill pathogens; however, excessive neutrophilic inflammation also leads to tissue injury and adverse effects. To discover novel therapeutic targets that modulate neutrophil migration, we performed a neutrophil-specific microRNA (miRNA) overexpression screen in zebrafish and identified 8 miRNAs as potent suppressors of neutrophil migration. Among those, miR-199 decreases neutrophil chemotaxis in zebrafish and human neutrophil-like cells. Intriguingly, in terminally differentiated neutrophils, miR-199 alters the cell cycle-related pathways and directly suppresses cyclin-dependent kinase 2 (Cdk2), whose known activity is restricted to cell cycle progression and cell differentiation. Inhibiting Cdk2, but not DNA replication, disrupts cell polarity and chemotaxis of zebrafish neutrophils without inducing cell death. Human neutrophil-like cells deficient in CDK2 fail to polarize and display altered signaling downstream of the formyl peptide receptor. Chemotaxis of primary human neutrophils is also reduced upon CDK2 inhibition. Furthermore, miR-199 overexpression or CDK2 inhibition significantly improves the outcome of lethal systemic inflammation challenges in zebrafish. Our results therefore reveal previously unknown functions of miR-199 and CDK2 in regulating neutrophil migration and provide directions in alleviating systemic inflammation.
Collapse
Affiliation(s)
- Alan Y Hsu
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Decheng Wang
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
- The Institute of Infection and Inflammation, Medical College of China Three Gorges University, 443002 Yichang, Hubei, People's Republic of China
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202
- Collaborative Core for Cancer Bioinformatics, Indiana University Simon Cancer Center, Indianapolis, IN 46202
| | - Justice Lu
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Ramizah Syahirah
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - David A Bennin
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53706
| | - David M Umulis
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202
- Collaborative Core for Cancer Bioinformatics, Indiana University Simon Cancer Center, Indianapolis, IN 46202
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Qing Deng
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907;
- Purdue Institute for Inflammation, Immunology, & Infectious Disease, Purdue University, West Lafayette, IN 47907
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907
| |
Collapse
|
28
|
Roca FJ, Whitworth LJ, Redmond S, Jones AA, Ramakrishnan L. TNF Induces Pathogenic Programmed Macrophage Necrosis in Tuberculosis through a Mitochondrial-Lysosomal-Endoplasmic Reticulum Circuit. Cell 2019; 178:1344-1361.e11. [PMID: 31474371 PMCID: PMC6736209 DOI: 10.1016/j.cell.2019.08.004] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/15/2019] [Accepted: 08/02/2019] [Indexed: 01/07/2023]
Abstract
Necrosis of infected macrophages constitutes a critical pathogenetic event in tuberculosis by releasing mycobacteria into the growth-permissive extracellular environment. In zebrafish infected with Mycobacterium marinum or Mycobacterium tuberculosis, excess tumor necrosis factor triggers programmed necrosis of infected macrophages through the production of mitochondrial reactive oxygen species (ROS) and the participation of cyclophilin D, a component of the mitochondrial permeability transition pore. Here, we show that this necrosis pathway is not mitochondrion-intrinsic but results from an inter-organellar circuit initiating and culminating in the mitochondrion. Mitochondrial ROS induce production of lysosomal ceramide that ultimately activates the cytosolic protein BAX. BAX promotes calcium flow from the endoplasmic reticulum into the mitochondrion through ryanodine receptors, and the resultant mitochondrial calcium overload triggers cyclophilin-D-mediated necrosis. We identify ryanodine receptors and plasma membrane L-type calcium channels as druggable targets to intercept mitochondrial calcium overload and necrosis of mycobacterium-infected zebrafish and human macrophages. TNF induces mitochondrial ROS to cause necrosis of mycobacterium-infected macrophages Mitochondrial ROS activate lysosomal enzymes that lead to BAX activation BAX activates ER ryanodine receptors to cause Ca2+ flow into the mitochondrion Drugs preventing mitochondrial Ca2+ overload prevent pathogenic macrophage necrosis in TB
Collapse
Affiliation(s)
- Francisco J Roca
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 OQH, UK.
| | - Laura J Whitworth
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 OQH, UK
| | - Sarah Redmond
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 OQH, UK; Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Ana A Jones
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 OQH, UK
| | - Lalita Ramakrishnan
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 OQH, UK; Department of Microbiology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
29
|
Black HD, Xu W, Hortle E, Robertson SI, Britton WJ, Kaur A, New EJ, Witting PK, Chami B, Oehlers SH. The cyclic nitroxide antioxidant 4-methoxy-TEMPO decreases mycobacterial burden in vivo through host and bacterial targets. Free Radic Biol Med 2019; 135:157-166. [PMID: 30878645 DOI: 10.1016/j.freeradbiomed.2019.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 03/07/2019] [Accepted: 03/09/2019] [Indexed: 12/22/2022]
Abstract
Tuberculosis is a chronic inflammatory disease caused by persistent infection with Mycobacterium tuberculosis. The rise of antibiotic resistant strains necessitates the design of novel treatments. Recent evidence shows that not only is M. tuberculosis highly resistant to oxidative killing, it also co-opts host oxidant production to induce phagocyte death facilitating bacterial dissemination. We have targeted this redox environment with the cyclic nitroxide derivative 4-methoxy-TEMPO (MetT) in the zebrafish-M. marinum infection model. MetT inhibited the production of mitochondrial ROS and decreased infection-induced cell death to aid containment of infection. We identify a second mechanism of action whereby stress conditions, including hypoxia, found in the infection microenvironment appear to sensitise M. marinum to killing by MetT both in vitro and in vivo. Together, our study demonstrates MetT inhibited the growth and dissemination of M. marinum through host and bacterial targets.
Collapse
Affiliation(s)
- Harrison D Black
- Centenary Institute, The University of Sydney, Australia; The University of Sydney, Discipline of Pathology Faculty of Medicine and Health, Australia
| | - Wenbo Xu
- Centenary Institute, The University of Sydney, Australia
| | - Elinor Hortle
- Centenary Institute, The University of Sydney, Australia; The University of Sydney, Central Clinical School Faculty of Medicine and Health and Marie Bashir Institute, Australia
| | | | - Warwick J Britton
- Centenary Institute, The University of Sydney, Australia; The University of Sydney, Central Clinical School Faculty of Medicine and Health and Marie Bashir Institute, Australia
| | - Amandeep Kaur
- The University of Sydney, School of Chemistry, Australia
| | | | - Paul K Witting
- The University of Sydney, Discipline of Pathology Faculty of Medicine and Health, Australia
| | - Belal Chami
- The University of Sydney, Discipline of Pathology Faculty of Medicine and Health, Australia
| | - Stefan H Oehlers
- Centenary Institute, The University of Sydney, Australia; The University of Sydney, Central Clinical School Faculty of Medicine and Health and Marie Bashir Institute, Australia.
| |
Collapse
|
30
|
Rosowski EE, Knox BP, Archambault LS, Huttenlocher A, Keller NP, Wheeler RT, Davis JM. The Zebrafish as a Model Host for Invasive Fungal Infections. J Fungi (Basel) 2018; 4:jof4040136. [PMID: 30551557 PMCID: PMC6308935 DOI: 10.3390/jof4040136] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 12/14/2022] Open
Abstract
The zebrafish has become a widely accepted model host for studies of infectious disease, including fungal infections. The species is genetically tractable, and the larvae are transparent and amenable to prolonged in vivo imaging and small molecule screening. The aim of this review is to provide a thorough introduction into the published studies of fungal infection in the zebrafish and the specific ways in which this model has benefited the field. In doing so, we hope to provide potential new zebrafish researchers with a snapshot of the current toolbox and prior results, while illustrating how the model has been used well and where the unfulfilled potential of this model can be found.
Collapse
Affiliation(s)
- Emily E Rosowski
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53716, USA.
| | - Benjamin P Knox
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53716, USA.
| | - Linda S Archambault
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA.
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53716, USA.
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53792, USA.
| | - Nancy P Keller
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53716, USA.
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Robert T Wheeler
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA.
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA.
| | - J Muse Davis
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
31
|
Trousil J, Ulmann V, Hrubý M. Fluorescence & bioluminescence in the quest for imaging, probing & analysis of mycobacterial infections. Future Microbiol 2018; 13:933-951. [PMID: 29893148 DOI: 10.2217/fmb-2017-0296] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mycobacterioses represent a global health problem and rapid diagnostic improvements are urgently required. Mycobacteria-specific fluorescence and bioluminescence phenomena have been found to be useful for a wide range of mycobacteria-focused research. Here, we present a critical survey of the most promising techniques in this field and the potential of new methods under investigation. These approaches include acid-fast staining, intrinsic fluorescence of the coenzyme F420, fluorogenic substrates (e.g., β-lactamase-sensitive coumpounds) and recombination of mycobacteria or mycobacteriophages. Probably the most interesting and emerging host-inspecting approach is in vivo imaging. Detection of fluorescence in vivo, however, is complicated by light scattering, light absorption, and autofluorescence, caused by the tissues. Despite this, many of these systems show promise as the foundations for improved rapid analysis and imaging of mycobacterial infections, both in vitro and in vivo.
Collapse
Affiliation(s)
- Jiří Trousil
- Department of Supramolecular Polymer Systems, Institute of Macromolecular Chemistry of the Academy of Sciences of the Czech Republic, Heyrovského náměstí 2, 162 06 Prague 6, Czech Republic.,Department of Analytical Chemistry, Charles University, Faculty of Science, Hlavova 8, 128 43 Praha 2, Czech Republic
| | - Vít Ulmann
- Laboratory for Mycobacterial Diagnostics and Tuberculosis, Regional Institute of Public Health in Ostrava, Partyzánské náměstí 7, 702 00 Ostrava, Czech Republic
| | - Martin Hrubý
- Department of Supramolecular Polymer Systems, Institute of Macromolecular Chemistry of the Academy of Sciences of the Czech Republic, Heyrovského náměstí 2, 162 06 Prague 6, Czech Republic
| |
Collapse
|
32
|
Spain M, Wong JKH, Nagalingam G, Batten JM, Hortle E, Oehlers SH, Jiang XF, Murage HE, Orford JT, Crisologo P, Triccas JA, Rutledge PJ, Todd MH. Antitubercular Bis-Substituted Cyclam Derivatives: Structure-Activity Relationships and in Vivo Studies. J Med Chem 2018; 61:3595-3608. [PMID: 29558124 DOI: 10.1021/acs.jmedchem.7b01569] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We recently reported the discovery of nontoxic cyclam-derived compounds that are active against drug-resistant Mycobacterium tuberculosis. In this paper we report exploration of the structure-activity relationship for this class of compounds, identifying several simpler compounds with comparable activity. The most promising compound identified, possessing significantly improved water solubility, displayed high levels of bacterial clearance in an in vivo zebrafish embryo model, suggesting this compound series has promise for in vivo treatment of tuberculosis.
Collapse
Affiliation(s)
- Malcolm Spain
- School of Chemistry , The University of Sydney , Sydney , NSW 2006 , Australia
| | - Joseph K-H Wong
- School of Chemistry , The University of Sydney , Sydney , NSW 2006 , Australia
| | - Gayathri Nagalingam
- Microbial Immunity and Pathogenesis Group, Department of Infectious Diseases and Immunology, Sydney Medical School , The University of Sydney , Sydney , NSW 2006 , Australia
| | - James M Batten
- School of Chemistry , The University of Sydney , Sydney , NSW 2006 , Australia
| | - Elinor Hortle
- Tuberculosis Research Program , Centenary Institute, Royal Prince Alfred Hospital , Missenden Road , Camperdown, Sydney , NSW 2050 , Australia
| | - Stefan H Oehlers
- Central Clinical School, Sydney Medical School , The University of Sydney , Sydney , NSW 2006 , Australia
| | - Xiao Fan Jiang
- School of Chemistry , The University of Sydney , Sydney , NSW 2006 , Australia
| | - Hasini E Murage
- School of Chemistry , The University of Sydney , Sydney , NSW 2006 , Australia
| | - Jack T Orford
- School of Chemistry , The University of Sydney , Sydney , NSW 2006 , Australia
| | - Patrick Crisologo
- School of Chemistry , The University of Sydney , Sydney , NSW 2006 , Australia
| | - James A Triccas
- Microbial Immunity and Pathogenesis Group, Department of Infectious Diseases and Immunology, Sydney Medical School , The University of Sydney , Sydney , NSW 2006 , Australia
| | - Peter J Rutledge
- School of Chemistry , The University of Sydney , Sydney , NSW 2006 , Australia
| | - Matthew H Todd
- School of Chemistry , The University of Sydney , Sydney , NSW 2006 , Australia
| |
Collapse
|
33
|
Clatworthy AE, Romano KP, Hung DT. Whole-organism phenotypic screening for anti-infectives promoting host health. Nat Chem Biol 2018; 14:331-341. [PMID: 29556098 PMCID: PMC9843822 DOI: 10.1038/s41589-018-0018-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 11/20/2017] [Indexed: 01/19/2023]
Abstract
To date, antibiotics have been identified on the basis of their ability to kill bacteria or inhibit their growth rather than directly for their capacity to improve clinical outcomes of infected patients. Although historically successful, this approach has led to the development of an antibiotic armamentarium that suffers from a number of shortcomings, including the inevitable emergence of resistance and, in certain infections, suboptimal efficacy leading to long treatment durations, infection recurrence, or high mortality and morbidity rates despite apparent bacterial sterilization. Conventional antibiotics fail to address the complexities of in vivo bacterial physiology and virulence, as well as the role of the host underlying the complex, dynamic interactions that cause disease. New interventions are needed, aimed at host outcome rather than microbiological cure. Here we review the role of screening models for cellular and whole-organism infection, including worms, flies, zebrafish, and mice, to identify novel therapeutic strategies and discuss their future implications.
Collapse
Affiliation(s)
- Anne E. Clatworthy
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Keith P. Romano
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA,Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Deborah T. Hung
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA,Department of Genetics, Harvard Medical School, Boston, MA, USA,Correspondence and requests for materials should be addressed to D.T.H.
| |
Collapse
|
34
|
Hsu AY, Gurol T, Sobreira TJP, Zhang S, Moore N, Cai C, Zhang ZY, Deng Q. Development and Characterization of an Endotoxemia Model in Zebra Fish. Front Immunol 2018; 9:607. [PMID: 29651289 PMCID: PMC5884884 DOI: 10.3389/fimmu.2018.00607] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/12/2018] [Indexed: 12/16/2022] Open
Abstract
Endotoxemia is a condition in which endotoxins enter the blood stream and cause systemic and sometimes lethal inflammation. Zebra fish provides a genetically tractable model organism for studying innate immunity, with additional advantages in live imaging and drug discovery. However, a bona fide endotoxemia model has not been established in zebra fish. Here, we have developed an acute endotoxemia model in zebra fish by injecting a single dose of LPS directly into the circulation. Hallmarks of human acute endotoxemia, including systemic inflammation, extensive tissue damage, circulation blockade, immune cell mobilization, and emergency hematopoiesis, were recapitulated in this model. Knocking out the adaptor protein Myd88 inhibited systemic inflammation and improved zebra fish survival. In addition, similar alternations of pathways with human acute endotoxemia were detected using global proteomic profiling and MetaCore™ pathway enrichment analysis. Furthermore, treating zebra fish with a protein tyrosine phosphatase nonreceptor type 11 (Shp2) inhibitor decreased systemic inflammation, immune mobilization, tissue damage, and improved survival in the endotoxemia model. Together, we have established and characterized the phenotypic and gene expression changes of a zebra fish endotoxemia model, which is amenable to genetic and pharmacological discoveries that can ultimately lead to a better mechanistic understanding of the dynamics and interplay of the innate immune system.
Collapse
Affiliation(s)
- Alan Y Hsu
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Theodore Gurol
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Tiago J P Sobreira
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States
| | - Sheng Zhang
- Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States.,Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Natalie Moore
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Chufan Cai
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Zhong-Yin Zhang
- Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States.,Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, United States.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Qing Deng
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, United States.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
35
|
Abstract
![]()
Current tuberculosis
(TB) drug development efforts are not sufficient
to end the global TB epidemic. Recent efforts have focused on the
development of whole-cell screening assays because biochemical, target-based
inhibitor screens during the last two decades have not delivered new
TB drugs. Mycobacterium tuberculosis (Mtb), the causative
agent of TB, encounters diverse microenvironments and can be found
in a variety of metabolic states in the human host. Due to the complexity
and heterogeneity of Mtb infection, no single model can fully recapitulate
the in vivo conditions in which Mtb is found in TB patients, and there
is no single “standard” screening condition to generate
hit compounds for TB drug development. However, current screening
assays have become more sophisticated as researchers attempt to mirror
the complexity of TB disease in the laboratory. In this review, we
describe efforts using surrogates and engineered strains of Mtb to
focus screens on specific targets. We explain model culture systems
ranging from carbon starvation to hypoxia, and combinations thereof,
designed to represent the microenvironment which Mtb encounters in
the human body. We outline ongoing efforts to model Mtb infection
in the lung granuloma. We assess these different models, their ability
to generate hit compounds, and needs for further TB drug development,
to provide direction for future TB drug discovery.
Collapse
Affiliation(s)
- Tianao Yuan
- Department of Chemistry, Stony Brook University , Stony Brook, New York 11794-3400, United States
| | - Nicole S Sampson
- Department of Chemistry, Stony Brook University , Stony Brook, New York 11794-3400, United States.,Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Research Centre at Stellenbosch University , Stellenbosch 7600, South Africa
| |
Collapse
|
36
|
Trofimov V, Costa-Gouveia J, Hoffmann E, Brodin P. Host-pathogen systems for early drug discovery against tuberculosis. Curr Opin Microbiol 2017; 39:143-151. [PMID: 29179041 DOI: 10.1016/j.mib.2017.11.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/17/2017] [Indexed: 12/21/2022]
Abstract
Tuberculosis (TB) is a global disease causing 1.8 million deaths each year. The appearance of drug-resistant strains raised the demand for new anti-mycobacterial drugs and therapies, because previously discovered antibiotics are shown to be inefficient. Moreover, the number of newly discovered drugs is not increasing in proportion to the emergence of drug resistance, which suggests that more optimized methodology and screening procedures are required including the incorporation of in vivo properties of TB infection. A way to improve efficacy of screening approaches is by introducing the use of different host-pathogen systems into primary screenings. These include whole cell-based screenings, zebrafish larvae-based screenings and the impact of artificial granuloma research on the drug discovery process. This review highlights current screening attempts and the identified molecular targets and summarizes findings of alternative, not fully explored host-pathogen systems for the characterization of anti-mycobacterial compounds.
Collapse
Affiliation(s)
- Valentin Trofimov
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, University Lille, Lille, France
| | - Joana Costa-Gouveia
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, University Lille, Lille, France
| | - Eik Hoffmann
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, University Lille, Lille, France
| | - Priscille Brodin
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, University Lille, Lille, France.
| |
Collapse
|
37
|
Boot M, Jim KK, Liu T, Commandeur S, Lu P, Verboom T, Lill H, Bitter W, Bald D. A fluorescence-based reporter for monitoring expression of mycobacterial cytochrome bd in response to antibacterials and during infection. Sci Rep 2017; 7:10665. [PMID: 28878275 PMCID: PMC5587683 DOI: 10.1038/s41598-017-10944-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 08/16/2017] [Indexed: 12/16/2022] Open
Abstract
Cytochrome bd is a component of the oxidative phosphorylation pathway in many Gram-positive and Gram-negative bacteria. Next to its role as a terminal oxidase in the respiratory chain this enzyme plays an important role as a survival factor in the bacterial stress response. In Mycobacterium tuberculosis and related mycobacterial strains, cytochrome bd is an important component of the defense system against antibacterial drugs. In this report we describe and evaluate an mCherry-based fluorescent reporter for detection of cytochrome bd expression in Mycobacterium marinum. Cytochrome bd was induced by mycolic acid biosynthesis inhibitors such as isoniazid and most prominently by drugs targeting oxidative phosphorylation. We observed no induction by inhibitors of protein-, DNA- or RNA-synthesis. The constructed expression reporter was suitable for monitoring mycobacterial cytochrome bd expression during mouse macrophage infection and in a zebrafish embryo infection model when using Mycobacterium marinum. Interestingly, in both these infection models cytochrome bd levels were considerably higher than during in vitro culturing of M. marinum. The expression reporter described here can be a valuable tool for elucidating the role of cytochrome bd as a survival factor.
Collapse
Affiliation(s)
- Maikel Boot
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Kin Ki Jim
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Ting Liu
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Susanna Commandeur
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Ping Lu
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Theo Verboom
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Holger Lill
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.,Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Dirk Bald
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.
| |
Collapse
|
38
|
Aspatwar A, Hammarén M, Koskinen S, Luukinen B, Barker H, Carta F, Supuran CT, Parikka M, Parkkila S. β-CA-specific inhibitor dithiocarbamate Fc14-584B: a novel antimycobacterial agent with potential to treat drug-resistant tuberculosis. J Enzyme Inhib Med Chem 2017. [PMID: 28629306 PMCID: PMC6445161 DOI: 10.1080/14756366.2017.1332056] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Inhibition of novel biological pathways in Mycobacterium tuberculosis (Mtb) creates the potential for alternative approaches for treating drug-resistant tuberculosis. In vitro studies have shown that dithiocarbamate-derived β-carbonic anhydrase (β-CA) inhibitors Fc14–594 A and Fc14–584B effectively inhibit the activity of Mtb β-CA enzymes. We screened the dithiocarbamates for toxicity, and studied the in vivo inhibitory effect of the least toxic inhibitor on M. marinum in a zebrafish model. In our toxicity screening, Fc14–584B emerged as the least toxic and showed minimal toxicity in 5-day-old larvae at 300 µM concentration. In vitro inhibition of M. marinum showed that both compounds inhibited growth at a concentration of 75 µM. In vivo inhibition studies using 300 µM Fc14–584B showed significant (p > .05) impairment of bacterial growth in zebrafish larvae at 6 days post infection. Our studies highlight the therapeutic potential of Fc14–584B as a β-CA inhibitor against Mtb, and that dithiocarbamate compounds may be developed into potent anti-tuberculosis drugs.
Collapse
Affiliation(s)
- Ashok Aspatwar
- a Faculty of Medicine and Life Sciences , University of Tampere , Tampere , Finland
| | - Milka Hammarén
- a Faculty of Medicine and Life Sciences , University of Tampere , Tampere , Finland
| | - Sanni Koskinen
- a Faculty of Medicine and Life Sciences , University of Tampere , Tampere , Finland
| | - Bruno Luukinen
- a Faculty of Medicine and Life Sciences , University of Tampere , Tampere , Finland
| | - Harlan Barker
- a Faculty of Medicine and Life Sciences , University of Tampere , Tampere , Finland
| | - Fabrizio Carta
- b Neurofarba Department , Sezione di Chimica Farmaceutica e Nutraceutica, Università degli Studi di Firenze , Sesto Fiorentino (Firenze) , Italy
| | - Claudiu T Supuran
- b Neurofarba Department , Sezione di Chimica Farmaceutica e Nutraceutica, Università degli Studi di Firenze , Sesto Fiorentino (Firenze) , Italy
| | - Mataleena Parikka
- a Faculty of Medicine and Life Sciences , University of Tampere , Tampere , Finland
| | - Seppo Parkkila
- a Faculty of Medicine and Life Sciences , University of Tampere , Tampere , Finland.,c Fimlab Ltd. and Tampere University Hospital , Tampere , Finland
| |
Collapse
|
39
|
Yao Y, Sun S, Fei F, Wang J, Wang Y, Zhang R, Wu J, Liu L, Liu X, Cui Z, Li Q, Yu M, Dang Y, Wang X. Screening in larval zebrafish reveals tissue-specific distribution of fifteen fluorescent compounds. Dis Model Mech 2017; 10:1155-1164. [PMID: 28754836 PMCID: PMC5611963 DOI: 10.1242/dmm.028811] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 07/14/2017] [Indexed: 01/05/2023] Open
Abstract
The zebrafish is a prominent vertebrate model for low-cost in vivo whole organism screening. In our recent screening of the distribution patterns of fluorescent compounds in live zebrafish larvae, fifteen compounds with tissue-specific distributions were identified. Several compounds were observed to accumulate in tissues where they were reported to induce side-effects, and compounds with similar structures tended to be enriched in the same tissues, with minor differences. In particular, we found three novel red fluorescent bone-staining dyes: purpurin, lucidin and 3-hydroxy-morindone; purpurin can effectively label bones in both larval and adult zebrafish, as well as in postnatal mice, without significantly affecting bone mass and density. Moreover, two structurally similar chemotherapeutic compounds, doxorubicin and epirubicin, were observed to have distinct distribution preferences in zebrafish. Epirubicin maintained a relatively higher concentration in the liver, and performed better in inhibiting hepatic hyperplasia caused by the over-expression of krasG12V In total, our study suggests that the transparent zebrafish larvae serve as valuable tools for identifying tissue-specific distributions of fluorescent compounds.
Collapse
Affiliation(s)
- Yuxiao Yao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shaoyang Sun
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Fei Fei
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jingjing Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Youhua Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ranran Zhang
- Institute of Reproduction and Development, Collaborative Innovation Center of Genetics and Development, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Jing Wu
- Deparment of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Lian Liu
- Institute of Reproduction and Development, Collaborative Innovation Center of Genetics and Development, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiuyun Liu
- Translational Medical Center for Development and Disease, Shanghai Key Laboratory of Birth Defects, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Zhaomeng Cui
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qiang Li
- Translational Medical Center for Development and Disease, Shanghai Key Laboratory of Birth Defects, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Min Yu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yongjun Dang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xu Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
40
|
Madigan CA, Cambier CJ, Kelly-Scumpia KM, Scumpia PO, Cheng TY, Zailaa J, Bloom BR, Moody DB, Smale ST, Sagasti A, Modlin RL, Ramakrishnan L. A Macrophage Response to Mycobacterium leprae Phenolic Glycolipid Initiates Nerve Damage in Leprosy. Cell 2017; 170:973-985.e10. [PMID: 28841420 PMCID: PMC5848073 DOI: 10.1016/j.cell.2017.07.030] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 05/13/2017] [Accepted: 07/19/2017] [Indexed: 12/21/2022]
Abstract
Mycobacterium leprae causes leprosy and is unique among mycobacterial diseases in producing peripheral neuropathy. This debilitating morbidity is attributed to axon demyelination resulting from direct interaction of the M. leprae-specific phenolic glycolipid 1 (PGL-1) with myelinating glia and their subsequent infection. Here, we use transparent zebrafish larvae to visualize the earliest events of M. leprae-induced nerve damage. We find that demyelination and axonal damage are not directly initiated by M. leprae but by infected macrophages that patrol axons; demyelination occurs in areas of intimate contact. PGL-1 confers this neurotoxic response on macrophages: macrophages infected with M. marinum-expressing PGL-1 also damage axons. PGL-1 induces nitric oxide synthase in infected macrophages, and the resultant increase in reactive nitrogen species damages axons by injuring their mitochondria and inducing demyelination. Our findings implicate the response of innate macrophages to M. leprae PGL-1 in initiating nerve damage in leprosy.
Collapse
Affiliation(s)
- Cressida A Madigan
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, University of Washington, Seattle, WA 98195, USA.
| | - C J Cambier
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Kindra M Kelly-Scumpia
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Philip O Scumpia
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tan-Yun Cheng
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph Zailaa
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Barry R Bloom
- Harvard School of Public Health, Boston, MA 02115, USA
| | - D Branch Moody
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stephen T Smale
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alvaro Sagasti
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Robert L Modlin
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lalita Ramakrishnan
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA; Department of Immunology, University of Washington, Seattle, WA 98195, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA; MRC Laboratory of Molecular Biology, Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 OQH, UK.
| |
Collapse
|
41
|
Cambier CJ, O'Leary SM, O'Sullivan MP, Keane J, Ramakrishnan L. Phenolic Glycolipid Facilitates Mycobacterial Escape from Microbicidal Tissue-Resident Macrophages. Immunity 2017; 47:552-565.e4. [PMID: 28844797 PMCID: PMC5610147 DOI: 10.1016/j.immuni.2017.08.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/27/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022]
Abstract
Mycobacterium tuberculosis (Mtb) enters the host in aerosol droplets deposited in lung alveoli, where the bacteria first encounter lung-resident alveolar macrophages. We studied the earliest mycobacterium-macrophage interactions in the optically transparent zebrafish. First-responding resident macrophages phagocytosed and eradicated infecting mycobacteria, suggesting that to establish a successful infection, mycobacteria must escape out of the initially infected resident macrophage into growth-permissive monocytes. We defined a critical role for mycobacterial membrane phenolic glycolipid (PGL) in engineering this transition. PGL activated the STING cytosolic sensing pathway in resident macrophages, inducing the production of the chemokine CCL2, which in turn recruited circulating CCR2+ monocytes toward infection. Transient fusion of infected macrophages with CCR2+ monocytes enabled bacterial transfer and subsequent dissemination, and interrupting this transfer so as to prolong mycobacterial sojourn in resident macrophages promoted clearing of infection. Human alveolar macrophages produced CCL2 in a PGL-dependent fashion following infection, arguing for the potential of PGL-blocking interventions or PGL-targeting vaccine strategies in the prevention of tuberculosis. Video Abstract
Microbicidal tissue-resident macrophages are first responders to mycobacteria Mycobacterial phenolic glycolipid induces macrophage CCL2 through STING activation CCL2 recruits mycobacterium-permissive monocytes to the tissue-resident macrophage Mycobacteria transfer from tissue macrophage to monocyte through a cell fusion event
Collapse
Affiliation(s)
- C J Cambier
- Department of Immunology, University of Washington, Seattle, WA 98195, USA; Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK; Department of Chemistry, Stanford University, Stanford, CT 94305, USA
| | - Seónadh M O'Leary
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland
| | - Mary P O'Sullivan
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland
| | - Joseph Keane
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland.
| | - Lalita Ramakrishnan
- Department of Immunology, University of Washington, Seattle, WA 98195, USA; Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK; Department of Microbiology, University of Washington, Seattle, WA 98195, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
42
|
Abstract
The interaction between the host and the pathogen is extremely complex and is affected by anatomical, physiological, and immunological diversity in the microenvironments, leading to phenotypic diversity of the pathogen. Phenotypic heterogeneity, defined as nongenetic variation observed in individual members of a clonal population, can have beneficial consequences especially in fluctuating stressful environmental conditions. This is all the more relevant in infections caused by Mycobacterium tuberculosis wherein the pathogen is able to survive and often establish a lifelong persistent infection in the host. Recent studies in tuberculosis patients and in animal models have documented the heterogeneous and diverging trajectories of individual lesions within a single host. Since the fate of the individual lesions appears to be determined by the local tissue environment rather than systemic response of the host, studying this heterogeneity is very relevant to ensure better control and complete eradication of the pathogen from individual lesions. The heterogeneous microenvironments greatly enhance M. tuberculosis heterogeneity influencing the growth rates, metabolic potential, stress responses, drug susceptibility, and eventual lesion resolution. Single-cell approaches such as time-lapse microscopy using microfluidic devices allow us to address cell-to-cell variations that are often lost in population-average measurements. In this review, we focus on some of the factors that could be considered as drivers of phenotypic heterogeneity in M. tuberculosis as well as highlight some of the techniques that are useful in addressing this issue.
Collapse
|
43
|
Szabo M, Svensson Akusjärvi S, Saxena A, Liu J, Chandrasekar G, Kitambi SS. Cell and small animal models for phenotypic drug discovery. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:1957-1967. [PMID: 28721015 PMCID: PMC5500539 DOI: 10.2147/dddt.s129447] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The phenotype-based drug discovery (PDD) approach is re-emerging as an alternative platform for drug discovery. This review provides an overview of the various model systems and technical advances in imaging and image analyses that strengthen the PDD platform. In PDD screens, compounds of therapeutic value are identified based on the phenotypic perturbations produced irrespective of target(s) or mechanism of action. In this article, examples of phenotypic changes that can be detected and quantified with relative ease in a cell-based setup are discussed. In addition, a higher order of PDD screening setup using small animal models is also explored. As PDD screens integrate physiology and multiple signaling mechanisms during the screening process, the identified hits have higher biomedical applicability. Taken together, this review highlights the advantages gained by adopting a PDD approach in drug discovery. Such a PDD platform can complement target-based systems that are currently in practice to accelerate drug discovery.
Collapse
Affiliation(s)
- Mihaly Szabo
- Department of Microbiology Tumor, and Cell Biology
| | | | - Ankur Saxena
- Department of Microbiology Tumor, and Cell Biology
| | - Jianping Liu
- Department of Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | | | | |
Collapse
|
44
|
Bernut A, Herrmann JL, Ordway D, Kremer L. The Diverse Cellular and Animal Models to Decipher the Physiopathological Traits of Mycobacterium abscessus Infection. Front Cell Infect Microbiol 2017; 7:100. [PMID: 28421165 PMCID: PMC5378707 DOI: 10.3389/fcimb.2017.00100] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/14/2017] [Indexed: 01/09/2023] Open
Abstract
Mycobacterium abscessus represents an important respiratory pathogen among the rapidly-growing non-tuberculous mycobacteria. Infections caused by M. abscessus are increasingly found in cystic fibrosis (CF) patients and are often refractory to antibiotic therapy. The underlying immunopathological mechanisms of pathogenesis remain largely unknown. A major reason for the poor advances in M. abscessus research has been a lack of adequate models to study the acute and chronic stages of the disease leading to delayed progress of evaluation of therapeutic efficacy of potentially active antibiotics. However, the recent development of cellular models led to new insights in the interplay between M. abscessus with host macrophages as well as with amoebae, proposed to represent the environmental host and reservoir for non-tuberculous mycobacteria. The zebrafish embryo has also appeared as a useful alternative to more traditional models as it recapitulates the vertebrate immune system and, due to its optical transparency, allows a spatio-temporal visualization of the infection process in a living animal. More sophisticated immunocompromised mice have also been exploited recently to dissect the immune and inflammatory responses to M. abscessus. Herein, we will discuss the limitations, advantages and potential offered by these various models to study the pathophysiology of M. abscessus infection and to assess the preclinical efficacy of compounds active against this emerging human pathogen.
Collapse
Affiliation(s)
- Audrey Bernut
- IRIM (ex-CPBS)-UMR 9004, Centre National de la Recherche Scientifique (CNRS), Infectious Disease Research Institute of Montpellier, Université de MontpellierMontpellier, France
| | - Jean-Louis Herrmann
- UMR 1173, Institut National de la Santé et de la Recherche Médicale, Université de Versailles Saint-Quentin-en-YvelinesMontigny-le-Bretonneux, France
| | - Diane Ordway
- Mycobacteria Research Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State UniversityFort Collins, CO, USA
| | - Laurent Kremer
- IRIM (ex-CPBS)-UMR 9004, Centre National de la Recherche Scientifique (CNRS), Infectious Disease Research Institute of Montpellier, Université de MontpellierMontpellier, France.,Institut National de la Santé et de la Recherche Médicale, IRIMMontpellier, France
| |
Collapse
|
45
|
Conrad WH, Osman MM, Shanahan JK, Chu F, Takaki KK, Cameron J, Hopkinson-Woolley D, Brosch R, Ramakrishnan L. Mycobacterial ESX-1 secretion system mediates host cell lysis through bacterium contact-dependent gross membrane disruptions. Proc Natl Acad Sci U S A 2017; 114:1371-1376. [PMID: 28119503 PMCID: PMC5307465 DOI: 10.1073/pnas.1620133114] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mycobacterium tuberculosis and Mycobacterium marinum are thought to exert virulence, in part, through their ability to lyse host cell membranes. The type VII secretion system ESX-1 [6-kDa early secretory antigenic target (ESAT-6) secretion system 1] is required for both virulence and host cell membrane lysis. Both activities are attributed to the pore-forming activity of the ESX-1-secreted substrate ESAT-6 because multiple studies have reported that recombinant ESAT-6 lyses eukaryotic membranes. We too find ESX-1 of M. tuberculosis and M. marinum lyses host cell membranes. However, we find that recombinant ESAT-6 does not lyse cell membranes. The lytic activity previously attributed to ESAT-6 is due to residual detergent in the preparations. We report here that ESX-1-dependent cell membrane lysis is contact dependent and accompanied by gross membrane disruptions rather than discrete pores. ESX-1-mediated lysis is also morphologically distinct from the contact-dependent lysis of other bacterial secretion systems. Our findings suggest redirection of research to understand the mechanism of ESX-1-mediated lysis.
Collapse
Affiliation(s)
- William H Conrad
- Department of Medicine, University of Cambridge, Cambridge CB2 0QH, United Kingdom
| | - Morwan M Osman
- Department of Medicine, University of Cambridge, Cambridge CB2 0QH, United Kingdom
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98105
| | - Jonathan K Shanahan
- Department of Medicine, University of Cambridge, Cambridge CB2 0QH, United Kingdom
- Wellcome Trust PhD Program in Infection, Immunity, and Inflammation, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Frances Chu
- Department of Microbiology, University of Washington, Seattle, WA 98105
| | - Kevin K Takaki
- Department of Medicine, University of Cambridge, Cambridge CB2 0QH, United Kingdom
| | - James Cameron
- Department of Microbiology, University of Washington, Seattle, WA 98105
| | | | - Roland Brosch
- Unit for Integrated Mycobacterial Pathogenomics, Institut Pasteur, 75015 Paris, France
| | - Lalita Ramakrishnan
- Department of Medicine, University of Cambridge, Cambridge CB2 0QH, United Kingdom;
- Department of Microbiology, University of Washington, Seattle, WA 98105
| |
Collapse
|
46
|
Barisch C, Soldati T. Mycobacterium marinum Degrades Both Triacylglycerols and Phospholipids from Its Dictyostelium Host to Synthesise Its Own Triacylglycerols and Generate Lipid Inclusions. PLoS Pathog 2017; 13:e1006095. [PMID: 28103313 PMCID: PMC5245797 DOI: 10.1371/journal.ppat.1006095] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/28/2016] [Indexed: 12/21/2022] Open
Abstract
During a tuberculosis infection and inside lipid-laden foamy macrophages, fatty acids (FAs) and sterols are the major energy and carbon source for Mycobacterium tuberculosis. Mycobacteria can be found both inside a vacuole and the cytosol, but how this impacts their access to lipids is not well appreciated. Lipid droplets (LDs) store FAs in form of triacylglycerols (TAGs) and are energy reservoirs of prokaryotes and eukaryotes. Using the Dictyostelium discoideum/Mycobacterium marinum infection model we showed that M. marinum accesses host LDs to build up its own intracytosolic lipid inclusions (ILIs). Here, we show that host LDs aggregate at regions of the bacteria that become exposed to the cytosol, and appear to coalesce on their hydrophobic surface leading to a transfer of diacylglycerol O-acyltransferase 2 (Dgat2)-GFP onto the bacteria. Dictyostelium knockout mutants for both Dgat enzymes are unable to generate LDs. Instead, the excess of exogenous FAs is esterified predominantly into phospholipids, inducing uncontrolled proliferation of the endoplasmic reticulum (ER). Strikingly, in absence of host LDs, M. marinum alternatively exploits these phospholipids, resulting in rapid reversal of ER-proliferation. In addition, the bacteria are unable to restrict their acquisition of lipids from the dgat1&2 double knockout leading to vast accumulation of ILIs. Recent data indicate that the presence of ILIs is one of the characteristics of dormant mycobacteria. During Dictyostelium infection, ILI formation in M. marinum is not accompanied by a significant change in intracellular growth and a reduction in metabolic activity, thus providing evidence that storage of neutral lipids does not necessarily induce dormancy.
Collapse
Affiliation(s)
- Caroline Barisch
- Department of Biochemistry, Science II, University of Geneva, 30 quai Ernest-Ansermet, Geneva, Switzerland
- * E-mail:
| | - Thierry Soldati
- Department of Biochemistry, Science II, University of Geneva, 30 quai Ernest-Ansermet, Geneva, Switzerland
| |
Collapse
|
47
|
Williams CH, Hong CC. Zebrafish small molecule screens: Taking the phenotypic plunge. Comput Struct Biotechnol J 2016; 14:350-356. [PMID: 27721960 PMCID: PMC5050293 DOI: 10.1016/j.csbj.2016.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/12/2016] [Accepted: 09/13/2016] [Indexed: 12/27/2022] Open
Abstract
Target based chemical screens are a mainstay of modern drug discovery, but the effectiveness of this reductionist approach is being questioned in light of declines in pharmaceutical R & D efficiency. In recent years, phenotypic screens have gained increasing acceptance as a complementary/alternative approach to early drug discovery. We discuss the various model organisms used in phenotypic screens, with particular focus on zebrafish, which has emerged as a leading model of in vivo phenotypic screens. Additionally, we anticipate therapeutic opportunities, particularly in orphan disease space, in the context of rapid advances in human Mendelian genetics, electronic health record (EHR)-enabled genome–phenome associations, and genome editing.
Collapse
Affiliation(s)
- Charles H Williams
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Charles C Hong
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| |
Collapse
|
48
|
Robertson AL, Ogryzko NV, Henry KM, Loynes CA, Foulkes MJ, Meloni MM, Wang X, Ford C, Jackson M, Ingham PW, Wilson HL, Farrow SN, Solari R, Flower RJ, Jones S, Whyte MKB, Renshaw SA. Identification of benzopyrone as a common structural feature in compounds with anti-inflammatory activity in a zebrafish phenotypic screen. Dis Model Mech 2016; 9:621-32. [PMID: 27079522 PMCID: PMC4920152 DOI: 10.1242/dmm.024935] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 04/04/2016] [Indexed: 12/14/2022] Open
Abstract
Neutrophils are essential for host defence and are recruited to sites of inflammation in response to tissue injury or infection. For inflammation to resolve, these cells must be cleared efficiently and in a controlled manner, either by apoptosis or reverse migration. If the inflammatory response is not well-regulated, persistent neutrophils can cause damage to host tissues and contribute to the pathogenesis of chronic inflammatory diseases, which respond poorly to current treatments. It is therefore important to develop drug discovery strategies that can identify new therapeutics specifically targeting neutrophils, either by promoting their clearance or by preventing their recruitment. Our recent in vivo chemical genetic screen for accelerators of inflammation resolution identified a subset of compounds sharing a common chemical signature, the bicyclic benzopyrone rings. Here, we further investigate the mechanisms of action of the most active of this chemical series, isopimpinellin, in our zebrafish model of neutrophilic inflammation. We found that this compound targets both the recruitment and resolution phases of the inflammatory response. Neutrophil migration towards a site of injury is reduced by isopimpinellin and this occurs as a result of PI3K inhibition. We also show that isopimpinellin induces neutrophil apoptosis to drive inflammation resolution in vivo using a new zebrafish reporter line detecting in vivo neutrophil caspase-3 activity and allowing quantification of flux through the apoptotic pathway in real time. Finally, our studies reveal that clinically available ‘cromones’ are structurally related to isopimpinellin and have previously undescribed pro-resolution activity in vivo. These findings could have implications for the therapeutic use of benzopyrones in inflammatory disease. Summary: Zebrafish inflammation screen identifies a new series of structurally related compounds with combined anti-inflammatory and pro-resolution activity, and reveals a previously unknown mechanism of action of clinical cromones.
Collapse
Affiliation(s)
- Anne L Robertson
- The Bateson Centre, University of Sheffield, Sheffield, S10 2TN, UK Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK Stem Cell Program and Division of Hematology/Oncology, Children's Hospital Boston, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, 02115 MA, USA
| | - Nikolay V Ogryzko
- The Bateson Centre, University of Sheffield, Sheffield, S10 2TN, UK Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Katherine M Henry
- The Bateson Centre, University of Sheffield, Sheffield, S10 2TN, UK Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Catherine A Loynes
- The Bateson Centre, University of Sheffield, Sheffield, S10 2TN, UK Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Matthew J Foulkes
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK Department of Chemistry, University of Sheffield, Sheffield, S10 2TN, UK
| | - Marco M Meloni
- Department of Chemistry, University of Sheffield, Sheffield, S10 2TN, UK
| | - Xingang Wang
- Wishtech Medical Technology, Weihai, Shandong, 264200, China
| | - Christopher Ford
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 9TX, UK MRC Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing, University of Liverpool, Liverpool, L7 9TX, UK
| | - Malcolm Jackson
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 9TX, UK MRC Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing, University of Liverpool, Liverpool, L7 9TX, UK
| | - Philip W Ingham
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Heather L Wilson
- The Bateson Centre, University of Sheffield, Sheffield, S10 2TN, UK Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Stuart N Farrow
- Institute of Human Development, University of Manchester, Manchester, M13 9PL, UK
| | - Roberto Solari
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, Norfolk Place, London, W2 1NY, UK
| | - Roderick J Flower
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Simon Jones
- Department of Chemistry, University of Sheffield, Sheffield, S10 2TN, UK
| | - Moira K B Whyte
- MRC/UoE Centre for Inflammation Research, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Stephen A Renshaw
- The Bateson Centre, University of Sheffield, Sheffield, S10 2TN, UK Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK MRC Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing, University of Liverpool, Liverpool, L7 9TX, UK
| |
Collapse
|
49
|
Abstract
Zebrafish embryos can be obtained for research purposes in large numbers at low cost and embryos develop externally in limited space, making them highly suitable for high-throughput cancer studies and drug screens. Non-invasive live imaging of various processes within the larvae is possible due to their transparency during development, and a multitude of available fluorescent transgenic reporter lines.To perform high-throughput studies, handling large amounts of embryos and larvae is required. With such high number of individuals, even minute tasks may become time-consuming and arduous. In this chapter, an overview is given of the developments in the automation of various steps of large scale zebrafish cancer research for discovering important cancer pathways and drugs for the treatment of human disease. The focus lies on various tools developed for cancer cell implantation, embryo handling and sorting, microfluidic systems for imaging and drug treatment, and image acquisition and analysis. Examples will be given of employment of these technologies within the fields of toxicology research and cancer research.
Collapse
|
50
|
Matty MA, Roca FJ, Cronan MR, Tobin DM. Adventures within the speckled band: heterogeneity, angiogenesis, and balanced inflammation in the tuberculous granuloma. Immunol Rev 2015; 264:276-87. [PMID: 25703566 DOI: 10.1111/imr.12273] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recent work in a variety of animal models, including mice, zebrafish, and macaques, as well as in humans, has led to a reassessment of several tenets of mycobacterial infection. In this review, we describe new findings about the composition and dynamics of the tuberculous granuloma, the central host structure in mycobacterial infection, as well as inflammatory mediators that drive a successful anti-microbial response on one hand and pathological inflammation on the other. We highlight granuloma heterogeneity that emerges in the context of infection, the functional consequences of angiogenesis in tuberculous granulomas, and data that balanced inflammation in humans, with a central role for tumor necrosis factor, appears to play a key role in optimal defense against mycobacterial infection. These findings have suggested new and specific host-directed therapies that await further clinical exploration.
Collapse
Affiliation(s)
- Molly A Matty
- Department of Molecular Genetics and Microbiology, Center for Host-Microbial Interactions, Duke University Medical Center, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | | | | | | |
Collapse
|