1
|
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
|
2
|
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
|
3
|
Huang X, Chu C, Shi C, Zhang J, Yan B, Shan F, Wang D, Shi Y, Peng C, Tang BZ. Seeing is believing: Efficiency evaluation of multifunctional ionic-dependent AIEgens for tuberculosis. Biomaterials 2023; 302:122301. [PMID: 37690379 DOI: 10.1016/j.biomaterials.2023.122301] [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: 06/01/2023] [Revised: 08/20/2023] [Accepted: 08/29/2023] [Indexed: 09/12/2023]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a significant public health threat with high rates of infection and mortality. Rapid and reliable theranostics of TB are essential to control transmission and shorten treatment duration. In this study, we report two cationic aggregation-inducing emission luminogens (AIEgens) named TTVP and TTPy, which have different functional charged moieties, to investigate their potential for simultaneous tracing and photodynamic therapy in TB infection. TTVP and TTPy exhibit intrinsic positive charges, excellent water solubility, and near-infrared (NIR) emission. Based on ionic-function relationships, TTVP, with more positive charges, demonstrates a stronger binding affinity to Mycobacterium marinum (M.m), (a close genetic relative of Mtb), compared to TTPy. Both TTVP and TTPy exhibit high efficiency in generating reactive oxygen species (ROS) when exposed to white light irradiation, enabling effective photodynamic killing of M.m in vitro. Additionally, we achieved long-term, real-time, noninvasive, continuous tracing, and evaluated therapeutic performance in vivo. Notably, TTVP outperformed TTPy in intracellular killing of M.m, suggesting a possible correlation between the labeling and photodynamic killing abilities of AIEgens. These findings provide valuable insights and a design basis for cationic AIEgens in TB research, offering potential advancements in TB theranostics.
Collapse
Affiliation(s)
- Xueni Huang
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, 200032, China; Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Chengshengze Chu
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Chunzi Shi
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Jiulong Zhang
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Bo Yan
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Fei Shan
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Dong Wang
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China.
| | - Yuxin Shi
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.
| | - Chen Peng
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China.
| |
Collapse
|
4
|
Di Paola D, Natale S, Gugliandolo E, Cordaro M, Crupi R, Siracusa R, D’Amico R, Fusco R, Impellizzeri D, Cuzzocrea S, Spanò N, Marino F, Peritore AF. Assessment of 2-Pentadecyl-2-oxazoline Role on Lipopolysaccharide-Induced Inflammation on Early Stage Development of Zebrafish ( Danio rerio). Life (Basel) 2022; 12:128. [PMID: 35054521 PMCID: PMC8781862 DOI: 10.3390/life12010128] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 02/06/2023] Open
Abstract
Lipopolysaccharide (LPS), or bacterial endotoxin, is an important virulence factor in several human and animal pathologies. Oxazoline of Palmitoylethanolamide (PEAOXA) has shown strong anti-inflammatory activity in several animal models. LPS was applied for 24 h to zebrafish embryos to induce inflammation, and then the anti-inflammatory action of PEAOXA was evaluated for the first time in the zebrafish model (Danio rerio). Different concentrations of PEAOXA were tested for toxicity on zebrafish embryonic development; only the highest concentration of 30 mg/L showed toxic effects. Quantitative RT-PCR was applied to detect Tumor necrosis factor-α, Interleukin 1β, 6, and 8, and members of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB). Exposure to LPS induced an increase in pro-inflammatory cytokines (tumor necrosis factor and interleukin 1, 6, and 8) in both gene and protein expression, as well as an increase of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) and the nuclear factor kappa light polypeptide enhancer in B-cells inhibitor (IκBα) gene expression. Furthermore, acute LPS exposure also induced an increase in tryptase release, related to mast cell activity, and in the production of apoptosis-related proteins (caspase 3, bax, and bcl-2). Treatment with PEAOXA 10 mg/L significantly counteracts LPS-induced inflammation in terms of cytokine expression and decreases tryptase release and the apoptosis pathway.
Collapse
Affiliation(s)
- Davide Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy; (D.D.P.); (S.N.); (R.S.); (R.D.); (R.F.); (D.I.); (F.M.); (A.F.P.)
| | - Sabrina Natale
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy; (D.D.P.); (S.N.); (R.S.); (R.D.); (R.F.); (D.I.); (F.M.); (A.F.P.)
| | - Enrico Gugliandolo
- Department of Veterinary Science, University of Messina, 98166 Messina, Italy; (E.G.); (R.C.)
| | - Marika Cordaro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98166 Messina, Italy;
| | - Rosalia Crupi
- Department of Veterinary Science, University of Messina, 98166 Messina, Italy; (E.G.); (R.C.)
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy; (D.D.P.); (S.N.); (R.S.); (R.D.); (R.F.); (D.I.); (F.M.); (A.F.P.)
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy; (D.D.P.); (S.N.); (R.S.); (R.D.); (R.F.); (D.I.); (F.M.); (A.F.P.)
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy; (D.D.P.); (S.N.); (R.S.); (R.D.); (R.F.); (D.I.); (F.M.); (A.F.P.)
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy; (D.D.P.); (S.N.); (R.S.); (R.D.); (R.F.); (D.I.); (F.M.); (A.F.P.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy; (D.D.P.); (S.N.); (R.S.); (R.D.); (R.F.); (D.I.); (F.M.); (A.F.P.)
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, Saint Louis, MO 63103, USA
| | - Nunziacarla Spanò
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98166 Messina, Italy;
| | - Fabio Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy; (D.D.P.); (S.N.); (R.S.); (R.D.); (R.F.); (D.I.); (F.M.); (A.F.P.)
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy; (D.D.P.); (S.N.); (R.S.); (R.D.); (R.F.); (D.I.); (F.M.); (A.F.P.)
| |
Collapse
|
5
|
Di Paola D, Capparucci F, Lanteri G, Cordaro M, Crupi R, Siracusa R, D'Amico R, Fusco R, Impellizzeri D, Cuzzocrea S, Spanò N, Gugliandolo E, Peritore AF. Combined Toxicity of Xenobiotics Bisphenol A and Heavy Metals on Zebrafish Embryos ( Danio rerio). TOXICS 2021; 9:toxics9120344. [PMID: 34941778 PMCID: PMC8706782 DOI: 10.3390/toxics9120344] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/05/2021] [Accepted: 11/26/2021] [Indexed: 12/19/2022]
Abstract
Environmental pollutants may cause adverse effects on the immune system of aquatic organisms. This study revealed that combination of environmental pollutants and Bisphenol A(BPA) could cause an acute inflammatory response in zebrafish larvae as shown by body alterations, which may imply a common immunotoxicity mechanism for most environmental pollutants. In the present study we evaluated the toxicity after co-exposure of BPA and Cd or Cr (III) in zebrafish embryos and larvae, and the oxidative stress pathway involved. Evaluation of lethal and developmental endpoints such as hatching, edema, malformations, abnormal heart rate and survival rate were evaluated after 96 h of exposure. Combination of BPA at 10 μM with Cd or Cr at 0.5 μM exposure induce malformations at 96 hpf in zebrafish larvae, as well as significantly increases oxidative stress and induce apoptosis on larvae. Our study suggested how environmental pollutant showed a synergistic effect at common not-effective doses, promoting decrease of antioxidant defense and contrasted fish development.
Collapse
Affiliation(s)
- Davide Di Paola
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| | - Fabiano Capparucci
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| | - Giovanni Lanteri
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98100 Messina, Italy
| | - Rosalia Crupi
- Department of Veterinary Science, University of Messina, 98100 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| | - Ramona D'Amico
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO 63108, USA
| | - Nunziacarla Spanò
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98100 Messina, Italy
| | - Enrico Gugliandolo
- Department of Veterinary Science, University of Messina, 98100 Messina, Italy
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| |
Collapse
|
6
|
Liu W, Zhang L, Sun S, Tang LS, He SM, Chen AQ, Yao LN, Ren DL. Cordycepin inhibits inflammatory responses through suppression of ERK activation in zebrafish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 124:104178. [PMID: 34157317 DOI: 10.1016/j.dci.2021.104178] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/03/2021] [Accepted: 06/16/2021] [Indexed: 06/13/2023]
Abstract
As a natural extract, cordycepin has been shown to play important regulatory roles in many life activities. In the study, the effects of cordycepin on inflammatory responses and the underlying mechanisms was explored using a zebrafish model. In the model of LPS-induced inflammation, cordycepin was found to significantly inhibited the expression of pro-inflammatory cytokines such as tnf-α, il-1β, il-6, and il-8. Using in vivo imaging model, cordycepin significantly inhibited fluorescent-labeled neutrophils migrating towards injury sites. Furthermore, results showed that the phosphorylation level of ERK protein dramatically decreased after cordycepin treatment. Meanwhile, the ERK inhibitor, PD0325901, significantly inhibited the expression of pro-inflammatory cytokines in LPS-induced inflammatory model and neutrophils migration in the caudal fin injury model. This study indicated the important roles of cordycepin in inhibiting LPS and injury-induced inflammation and preliminarily explained the role of ERK protein in this process.
Collapse
Affiliation(s)
- Wei Liu
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036, China
| | - Ling Zhang
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036, China
| | - Shuo Sun
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036, China
| | - Long-Sheng Tang
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036, China
| | - Shi-Min He
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036, China
| | - An-Qi Chen
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036, China
| | - Li-Na Yao
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036, China
| | - Da-Long Ren
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China.
| |
Collapse
|
7
|
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: 4.3] [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
|
8
|
Xie Y, Xie J, Meijer AH, Schaaf MJM. Glucocorticoid-Induced Exacerbation of Mycobacterial Infection Is Associated With a Reduced Phagocytic Capacity of Macrophages. Front Immunol 2021; 12:618569. [PMID: 34046029 PMCID: PMC8148013 DOI: 10.3389/fimmu.2021.618569] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 04/13/2021] [Indexed: 01/20/2023] Open
Abstract
Glucocorticoids are effective drugs for treating immune-related diseases, but prolonged therapy is associated with an increased risk of various infectious diseases, including tuberculosis. In this study, we have used a larval zebrafish model for tuberculosis, based on Mycobacterium marinum (Mm) infection, to study the effect of glucocorticoids. Our results show that the synthetic glucocorticoid beclomethasone increases the bacterial burden and the dissemination of a systemic Mm infection. The exacerbated Mm infection was associated with a decreased phagocytic activity of macrophages, higher percentages of extracellular bacteria, and a reduced rate of infected cell death, whereas the bactericidal capacity of the macrophages was not affected. The inhibited phagocytic capacity of macrophages was associated with suppression of the transcription of genes involved in phagocytosis in these cells. The decreased bacterial phagocytosis by macrophages was not specific for Mm, since it was also observed upon infection with Salmonella Typhimurium. In conclusion, our results show that glucocorticoids inhibit the phagocytic activity of macrophages, which may increase the severity of bacterial infections like tuberculosis.
Collapse
Affiliation(s)
- Yufei Xie
- Institute of Biology, Leiden University, Leiden, Netherlands
| | - Jiajun Xie
- Institute of Biology, Leiden University, Leiden, Netherlands
| | | | | |
Collapse
|
9
|
Baranyai Z, Soria‐Carrera H, Alleva M, Millán‐Placer AC, Lucía A, Martín‐Rapún R, Aínsa JA, la Fuente JM. Nanotechnology‐Based Targeted Drug Delivery: An Emerging Tool to Overcome Tuberculosis. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000113] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Zsuzsa Baranyai
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
| | - Héctor Soria‐Carrera
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- Biomateriales y Nanomedicina (CIBER‐BBN), Instituto de Salud Carlos III CIBER de Bioingeniería Madrid 28029 Spain
| | - Maria Alleva
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
| | - Ana C. Millán‐Placer
- Departamento de Microbiología, Facultad de Medicina Universidad de Zaragoza C/ Domingo Miral s/n Zaragoza 50009 Spain
- Instituto de Investigación Sanitaria Aragón (IIS‐Aragón) Zaragoza 50009 Spain
| | - Ainhoa Lucía
- Departamento de Microbiología, Facultad de Medicina Universidad de Zaragoza C/ Domingo Miral s/n Zaragoza 50009 Spain
- Instituto de Investigación Sanitaria Aragón (IIS‐Aragón) Zaragoza 50009 Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- CIBER de Enfermedades Respiratorias (CIBERES) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Rafael Martín‐Rapún
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- Departamento de Química Orgánica Facultad de Ciencias Universidad de Zaragoza Zaragoza 50009 Spain
- Biomateriales y Nanomedicina (CIBER‐BBN), Instituto de Salud Carlos III CIBER de Bioingeniería Madrid 28029 Spain
| | - José A. Aínsa
- Departamento de Microbiología, Facultad de Medicina Universidad de Zaragoza C/ Domingo Miral s/n Zaragoza 50009 Spain
- Instituto de Investigación Sanitaria Aragón (IIS‐Aragón) Zaragoza 50009 Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- CIBER de Enfermedades Respiratorias (CIBERES) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Jesús M. la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- Biomateriales y Nanomedicina (CIBER‐BBN), Instituto de Salud Carlos III CIBER de Bioingeniería Madrid 28029 Spain
| |
Collapse
|
10
|
Tăbăran AF, Matea CT, Mocan T, Tăbăran A, Mihaiu M, Iancu C, Mocan L. Silver Nanoparticles for the Therapy of Tuberculosis. Int J Nanomedicine 2020; 15:2231-2258. [PMID: 32280217 PMCID: PMC7127828 DOI: 10.2147/ijn.s241183] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/15/2020] [Indexed: 12/12/2022] Open
Abstract
Rapid emergence of aggressive, multidrug-resistant Mycobacteria strain represents the main cause of the current antimycobacterial-drug crisis and status of tuberculosis (TB) as a major global health problem. The relatively low-output of newly approved antibiotics contributes to the current orientation of research towards alternative antibacterial molecules such as advanced materials. Nanotechnology and nanoparticle research offers several exciting new-concepts and strategies which may prove to be valuable tools in improving the TB therapy. A new paradigm in antituberculous therapy using silver nanoparticles has the potential to overcome the medical limitations imposed in TB treatment by the drug resistance which is commonly reported for most of the current organic antibiotics. There is no doubt that AgNPs are promising future therapeutics for the medication of mycobacterial-induced diseases but the viability of this complementary strategy depends on overcoming several critical therapeutic issues as, poor delivery, variable intramacrophagic antimycobacterial efficiency, and residual toxicity. In this paper, we provide an overview of the pathology of mycobacterial-induced diseases, andhighlight the advantages and limitations of silver nanoparticles (AgNPs) in TB treatment.
Collapse
Affiliation(s)
- Alexandru-Flaviu Tăbăran
- Department of Pathology, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
- Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania
| | - Cristian Tudor Matea
- Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania
| | - Teodora Mocan
- Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania
- Department of Physiology, University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandra Tăbăran
- Department of Public Health and Food Hygiene, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Marian Mihaiu
- Department of Public Health and Food Hygiene, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Cornel Iancu
- Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania
- Third Surgery Department, University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lucian Mocan
- Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania
- Department of Physiology, University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
11
|
Sommer F, Torraca V, Meijer AH. Chemokine Receptors and Phagocyte Biology in Zebrafish. Front Immunol 2020; 11:325. [PMID: 32161595 PMCID: PMC7053378 DOI: 10.3389/fimmu.2020.00325] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 02/10/2020] [Indexed: 12/11/2022] Open
Abstract
Phagocytes are highly motile immune cells that ingest and clear microbial invaders, harmful substances, and dying cells. Their function is critically dependent on the expression of chemokine receptors, a class of G-protein-coupled receptors (GPCRs). Chemokine receptors coordinate the recruitment of phagocytes and other immune cells to sites of infection and damage, modulate inflammatory and wound healing responses, and direct cell differentiation, proliferation, and polarization. Besides, a structurally diverse group of atypical chemokine receptors (ACKRs) are unable to signal in G-protein-dependent fashion themselves but can shape chemokine gradients by fine-tuning the activity of conventional chemokine receptors. The optically transparent zebrafish embryos and larvae provide a powerful in vivo system to visualize phagocytes during development and study them as key elements of the immune response in real-time. In this review, we discuss how the zebrafish model has furthered our understanding of the role of two main classes of chemokine receptors, the CC and CXC subtypes, in phagocyte biology. We address the roles of the receptors in the migratory properties of phagocytes in zebrafish models for cancer, infectious disease, and inflammation. We illustrate how studies in zebrafish enable visualizing the contribution of chemokine receptors and ACKRs in shaping self-generated chemokine gradients of migrating cells. Taking the functional antagonism between two paralogs of the CXCR3 family as an example, we discuss how the duplication of chemokine receptor genes in zebrafish poses challenges, but also provides opportunities to study sub-functionalization or loss-of-function events. We emphasize how the zebrafish model has been instrumental to prove that the major determinant for the functional outcome of a chemokine receptor-ligand interaction is the cell-type expressing the receptor. Finally, we highlight relevant homologies and analogies between mammalian and zebrafish phagocyte function and discuss the potential of zebrafish models to further advance our understanding of chemokine receptors in innate immunity and disease.
Collapse
Affiliation(s)
- Frida Sommer
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Vincenzo Torraca
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | | |
Collapse
|
12
|
Hu W, Yang S, Shimada Y, Münch M, Marín-Juez R, Meijer AH, Spaink HP. Infection and RNA-seq analysis of a zebrafish tlr2 mutant shows a broad function of this toll-like receptor in transcriptional and metabolic control and defense to Mycobacterium marinum infection. BMC Genomics 2019; 20:878. [PMID: 31747871 PMCID: PMC6869251 DOI: 10.1186/s12864-019-6265-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
Background The function of Toll-like receptor 2 (TLR2) in host defense against pathogens, especially Mycobacterium tuberculosis (Mtb) is poorly understood. To investigate the role of TLR2 during mycobacterial infection, we analyzed the response of tlr2 zebrafish mutant larvae to infection with Mycobacterium marinum (Mm), a close relative to Mtb, as a model for tuberculosis. We measured infection phenotypes and transcriptome responses using RNA deep sequencing in mutant and control larvae. Results tlr2 mutant embryos at 2 dpf do not show differences in numbers of macrophages and neutrophils compared to control embryos. However, we found substantial changes in gene expression in these mutants, particularly in metabolic pathways, when compared with the heterozygote tlr2+/− control. At 4 days after Mm infection, the total bacterial burden and the presence of extracellular bacteria were higher in tlr2−/− larvae than in tlr2+/−, or tlr2+/+ larvae, whereas granuloma numbers were reduced, showing a function of Tlr2 in zebrafish host defense. RNAseq analysis of infected tlr2−/− versus tlr2+/− shows that the number of up-regulated and down-regulated genes in response to infection was greatly diminished in tlr2 mutants by at least 2 fold and 10 fold, respectively. Analysis of the transcriptome data and qPCR validation shows that Mm infection of tlr2 mutants leads to decreased mRNA levels of genes involved in inflammation and immune responses, including il1b, tnfb, cxcl11aa/ac, fosl1a, and cebpb. Furthermore, RNAseq analyses revealed that the expression of genes for Maf family transcription factors, vitamin D receptors, and Dicps proteins is altered in tlr2 mutants with or without infection. In addition, the data indicate a function of Tlr2 in the control of induction of cytokines and chemokines, such as the CXCR3-CXCL11 signaling axis. Conclusion The transcriptome and infection burden analyses show a function of Tlr2 as a protective factor against mycobacteria. Transcriptome analysis revealed tlr2-specific pathways involved in Mm infection, which are related to responses to Mtb infection in human macrophages. Considering its dominant function in control of transcriptional processes that govern defense responses and metabolism, the TLR2 protein can be expected to be also of importance for other infectious diseases and interactions with the microbiome.
Collapse
Affiliation(s)
- Wanbin Hu
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands
| | - Shuxin Yang
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.,Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yasuhito Shimada
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.,Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Magnus Münch
- Mathematical Institute, Leiden University, Leiden, the Netherlands.,Department of Epidemiology & Biostatistics, Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Rubén Marín-Juez
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.,Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany
| | - Annemarie H Meijer
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands
| | - Herman P Spaink
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.
| |
Collapse
|
13
|
Sommer F, Torraca V, Kamel SM, Lombardi A, Meijer AH. Frontline Science: Antagonism between regular and atypical Cxcr3 receptors regulates macrophage migration during infection and injury in zebrafish. J Leukoc Biol 2019; 107:185-203. [PMID: 31529512 PMCID: PMC7028096 DOI: 10.1002/jlb.2hi0119-006r] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 07/11/2019] [Accepted: 09/04/2019] [Indexed: 12/17/2022] Open
Abstract
The CXCR3‐CXCL11 chemokine‐signaling axis plays an essential role in infection and inflammation by orchestrating leukocyte trafficking in human and animal models, including zebrafish. Atypical chemokine receptors (ACKRs) play a fundamental regulatory function in signaling networks by shaping chemokine gradients through their ligand scavenging function, while being unable to signal in the classic G‐protein‐dependent manner. Two copies of the CXCR3 gene in zebrafish, cxcr3.2 and cxcr3.3, are expressed on macrophages and share a highly conserved ligand‐binding site. However, Cxcr3.3 has structural characteristics of ACKRs indicative of a ligand‐scavenging role. In contrast, we previously showed that Cxcr3.2 is an active CXCR3 receptor because it is required for macrophage motility and recruitment to sites of mycobacterial infection. In this study, we generated a cxcr3.3 CRISPR‐mutant to functionally dissect the antagonistic interplay among the cxcr3 paralogs in the immune response. We observed that cxcr3.3 mutants are more susceptible to mycobacterial infection, whereas cxcr3.2 mutants are more resistant. Furthermore, macrophages in the cxcr3.3 mutant are more motile, show higher activation status, and are recruited more efficiently to sites of infection or injury. Our results suggest that Cxcr3.3 is an ACKR that regulates the activity of Cxcr3.2 by scavenging common ligands and that silencing the scavenging function of Cxcr3.3 results in an exacerbated Cxcr3.2 signaling. In human, splice variants of CXCR3 have antagonistic functions and CXCR3 ligands also interact with ACKRs. Therefore, in zebrafish, an analogous regulatory mechanism appears to have evolved after the cxcr3 gene duplication event, through diversification of conventional and atypical receptor variants.
Collapse
Affiliation(s)
- Frida Sommer
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Vincenzo Torraca
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Sarah M Kamel
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Amber Lombardi
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | | |
Collapse
|
14
|
Chakraborty P, Kumar A. The extracellular matrix of mycobacterial biofilms: could we shorten the treatment of mycobacterial infections? MICROBIAL CELL 2019; 6:105-122. [PMID: 30740456 PMCID: PMC6364259 DOI: 10.15698/mic2019.02.667] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
A number of non-tuberculous mycobacterium species are opportunistic pathogens and ubiquitously form biofilms. These infections are often recalcitrant to treatment and require therapy with multiple drugs for long duration. The biofilm resident bacteria also display phenotypic drug tolerance and thus it has been hypothesized that the drug unresponsiveness in vivo could be due to formation of biofilms inside the host. We have discussed the biofilms of several pathogenic non-tuberculous mycobacterium (NTM) species in context to the in vivo pathologies. Besides pathogenic NTMs, Mycobacterium smegmatis is often used as a model organism for understanding mycobacterial physiology and has been studied extensively for understanding the mycobacterial biofilms. A number of components of the mycobacterial cell wall such as glycopeptidolipids, short chain mycolic acids, monomeromycolyl diacylglycerol, etc. have been shown to play an important role in formation of pellicle biofilms. It shall be noted that these components impart a hydrophobic character to the mycobacterial cell surface that facilitates cell to cell interaction. However, these components are not necessarily the constituents of the extracellular matrix of mycobacterial biofilms. In the end, we have described the biofilms of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis. Three models of Mtb biofilm formation have been proposed to study the factors regulating biofilm formation, the physiology of the resident bacteria, and the nature of the biomaterial that holds these bacterial masses together. These models include pellicle biofilms formed at the liquid-air interface of cultures, leukocyte lysate-induced biofilms, and thiol reductive stressinduced biofilms. All the three models offer their own advantages in the study of Mtb biofilms. Interestingly, lipids (mainly keto-mycolic acids) are proposed to be the primary component of extracellular polymeric substance (EPS) in the pellicle biofilm, whereas the leukocyte lysate-induced and thiol reductive stress-induced biofilms possess polysaccharides as the primary component of EPS. Both models also contain extracellular DNA in the EPS. Interestingly, thiol reductive stressinduced Mtb biofilms are held together by cellulose and yet unidentified structural proteins. We believe that a better understanding of the EPS of Mtb biofilms and the physiology of the resident bacteria will facilitate the development of shorter regimen for TB treatment.
Collapse
Affiliation(s)
- Poushali Chakraborty
- Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, India 160036
| | - Ashwani Kumar
- Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, India 160036.,CSIR-Academy of Scientific & Innovative Research (AcSIR), Council of Scientific & Industrial Research, New Delhi-110001
| |
Collapse
|
15
|
Luukinen H, Hammarén MM, Vanha-Aho LM, Parikka M. Modeling Tuberculosis in Mycobacterium marinum Infected Adult Zebrafish. J Vis Exp 2018. [PMID: 30346391 DOI: 10.3791/58299] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mycobacterium tuberculosis is currently the deadliest human pathogen causing 1.7 million deaths and 10.4 million infections every year. Exposure to this bacterium causes a wide disease spectrum in humans ranging from a sterilized infection to an actively progressing deadly disease. The most common form is the latent tuberculosis, which is asymptomatic, but has the potential to reactivate into a fulminant disease. Adult zebrafish and its natural pathogen Mycobacterium marinum have recently proven to be an applicable model to study the wide disease spectrum of tuberculosis. Importantly, spontaneous latency and reactivation as well as adaptive immune responses in the context of mycobacterial infection can be studied in this model. In this article, we describe methods for the experimental infection of adult zebrafish, the collection of internal organs for the extraction of nucleic acids for the measurement of mycobacterial loads and host immune responses by quantitative PCR. The in-house-developed, M. marinum-specific qPCR assay is more sensitive than the traditional plating methods as it also detects DNA from non-dividing, dormant or recently dead mycobacteria. As both DNA and RNA are extracted from the same individual, it is possible to study the relationships between the diseased state, and the host and pathogen gene-expression. The adult zebrafish model for tuberculosis thus presents itself as a highly applicable, non-mammalian in vivo system to study host-pathogen interactions.
Collapse
Affiliation(s)
- Hanna Luukinen
- Faculty of Medicine and Life Sciences, University of Tampere;
| | | | | | - Mataleena Parikka
- Faculty of Medicine and Life Sciences, University of Tampere; Oral and Maxillofacial Unit, Tampere University Hospital
| |
Collapse
|
16
|
van Leeuwen LM, Boot M, Kuijl C, Picavet DI, van Stempvoort G, van der Pol SM, de Vries HE, van der Wel NN, van der Kuip M, van Furth AM, van der Sar AM, Bitter W. Mycobacteria employ two different mechanisms to cross the blood-brain barrier. Cell Microbiol 2018; 20:e12858. [PMID: 29749044 PMCID: PMC6175424 DOI: 10.1111/cmi.12858] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/27/2018] [Accepted: 04/23/2018] [Indexed: 12/16/2022]
Abstract
Central nervous system (CNS) infection by Mycobacterium tuberculosis is one of the most devastating complications of tuberculosis, in particular in early childhood. In order to induce CNS infection, M. tuberculosis needs to cross specialised barriers protecting the brain. How M. tuberculosis crosses the blood-brain barrier (BBB) and enters the CNS is not well understood. Here, we use transparent zebrafish larvae and the closely related pathogen Mycobacterium marinum to answer this question. We show that in the early stages of development, mycobacteria rapidly infect brain tissue, either as free mycobacteria or within circulating macrophages. After the formation of a functionally intact BBB, the infiltration of brain tissue by infected macrophages is delayed, but not blocked, suggesting that crossing the BBB via phagocytic cells is one of the mechanisms used by mycobacteria to invade the CNS. Interestingly, depletion of phagocytic cells did not prevent M. marinum from infecting the brain tissue, indicating that free mycobacteria can independently cause brain infection. Detailed analysis showed that mycobacteria are able to cause vasculitis by extracellular outgrowth in the smaller blood vessels and by infecting endothelial cells. Importantly, we could show that this second mechanism is an active process that depends on an intact ESX-1 secretion system, which extends the role of ESX-1 secretion beyond the macrophage infection cycle.
Collapse
Affiliation(s)
- Lisanne M. van Leeuwen
- Medical Microbiology and Infection ControlVU Medical CenterAmsterdamThe Netherlands
- Paediatric Infectious Diseases and ImmunologyVU Medical CenterAmsterdamThe Netherlands
| | - Maikel Boot
- Medical Microbiology and Infection ControlVU Medical CenterAmsterdamThe Netherlands
| | - Coen Kuijl
- Medical Microbiology and Infection ControlVU Medical CenterAmsterdamThe Netherlands
| | - Daisy I. Picavet
- Cell Biology and Histology, Electron Microscopy Centre AmsterdamAcademic Medical CentreAmsterdamThe Netherlands
| | - Gunny van Stempvoort
- Medical Microbiology and Infection ControlVU Medical CenterAmsterdamThe Netherlands
| | - Susanne M.A. van der Pol
- Molecular Cell Biology and Immunology, Amsterdam NeuroscienceVU Medical CenterAmsterdamThe Netherlands
| | - Helga E. de Vries
- Molecular Cell Biology and Immunology, Amsterdam NeuroscienceVU Medical CenterAmsterdamThe Netherlands
| | - Nicole N. van der Wel
- Cell Biology and Histology, Electron Microscopy Centre AmsterdamAcademic Medical CentreAmsterdamThe Netherlands
| | - Martijn van der Kuip
- Paediatric Infectious Diseases and ImmunologyVU Medical CenterAmsterdamThe Netherlands
| | | | | | - Wilbert Bitter
- Medical Microbiology and Infection ControlVU Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
17
|
Vyas SP, Goswami R. Striking the right immunological balance prevents progression of tuberculosis. Inflamm Res 2017; 66:1031-1056. [PMID: 28711989 DOI: 10.1007/s00011-017-1081-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/03/2017] [Accepted: 07/07/2017] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Tuberculosis (TB) caused by infection with Mycobacterium tuberculosis (Mtb) is a major burden for human health worldwide. Current standard treatments for TB require prolonged administration of antimycobacterial drugs leading to exaggerated inflammation and tissue damage. This can result in the reactivation of latent TB culminating in TB progression. Thus, there is an unmet need to develop therapies that would shorten the duration of anti-TB treatment and to induce optimal protective immune responses to control the spread of mycobacterial infection with minimal lung pathology. FINDINGS Granulomata is the hallmark structure formed by the organized accumulation of immune cells including macrophages, natural killer cells, dendritic cells, neutrophils, T cells, and B cells to the site of Mtb infection. It safeguards the host by containing Mtb in latent form. However, granulomata can undergo caseation and contribute to the reactivation of latent TB, if the immune responses developed to fight mycobacterial infection are not properly controlled. Thus, an optimal balance between innate and adaptive immune cells might play a vital role in containing mycobacteria in latent form for prolonged periods and prevent the spread of Mtb infection from one individual to another. CONCLUSION Optimal and well-regulated immune responses against Mycobacterium tuberculosis may help to prevent the reactivation of latent TB. Moreover, therapies targeting balanced immune responses could help to improve treatment outcomes among latently infected TB patients and thereby limit the dissemination of mycobacterial infection.
Collapse
Affiliation(s)
| | - Ritobrata Goswami
- School of Bio Science, IIT Kharagpur, Kharagpur, West Bengal, 721302, India.
| |
Collapse
|
18
|
Abstract
The development of the zebrafish (Danio rerio) infectious disease model has provided new insights and information into pathogenesis. Many of these new discoveries would not have been possible using a typical mammalian model. The advantages of using this model are many and in the last 15 years the model has been exploited for the analysis of many different pathogens. Here, we describe in detail how to perform a bacterial infection using either the adult zebrafish or zebrafish larvae using microinjection. Multiple methods of analysis are described that can be used to address specific questions pertaining to disease progression and the interactions with the immune system.
Collapse
Affiliation(s)
- Melody N Neely
- Department of Biology, Texas Woman's University, 1200 Frame St., P.O. Box 425799, Denton, TX, 76204, USA.
| |
Collapse
|
19
|
Saraceni PR, Romero A, Figueras A, Novoa B. Establishment of Infection Models in Zebrafish Larvae (Danio rerio) to Study the Pathogenesis of Aeromonas hydrophila. Front Microbiol 2016; 7:1219. [PMID: 27540375 PMCID: PMC4972827 DOI: 10.3389/fmicb.2016.01219] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 07/21/2016] [Indexed: 12/12/2022] Open
Abstract
Aeromonas hydrophila is a Gram-negative opportunistic pathogen of fish and terrestrial animals. In humans, A. hydrophila mainly causes gastroenteritis, septicaemia, and tissue infections. The mechanisms of infection, the main virulence factors and the host immune response triggered by A. hydrophila have been studied in detail using murine models and adult fish. However, the great limitation of studying adult animals is that the animal must be sacrificed and its tissues/organs extracted, which prevents the study of the infectious processes in the whole living animal. Zebrafish larvae are being used for the analysis of several infectious diseases, but their use for studying the pathogenesis of A. hydrophila has never been explored. The great advantage of zebrafish larvae is their transparency during the first week after fertilization, which allows detailed descriptions of the infectious processes using in vivo imaging techniques such as differential interferential contrast (DIC) and fluorescence microscopy. Moreover, the availability of fluorescent pathogens and transgenic reporter zebrafish lines expressing fluorescent immune cells, immune marker genes or cytokines/chemokines allows the host-pathogen interactions to be characterized. The present study explores the suitability of zebrafish larvae to study the pathogenesis of A. hydrophila and the interaction mechanisms between the bacterium and the innate immune responses through an infection model using different routes for infection. We used an early-embryo infection model at 3 days post-fertilization (dpf) through the microinjection of A. hydrophila into the duct of Cuvier, caudal vein, notochord, or muscle and two bath infection models using 4 dpf healthy and injured larvae. The latter resembled the natural conditions under which A. hydrophila produces infectious diseases in animals. We compared the cellular processes after infection in each anatomical site by confocal fluorescence imaging and determined the implication of inflammatory immune genes by measuring gene expression by qPCR.
Collapse
Affiliation(s)
| | | | | | - Beatriz Novoa
- Immunology and Genomics, Institute of Marine Research (IIM) – Consejo Superior de Investigaciones Científicas (CSIC), VigoSpain
| |
Collapse
|
20
|
Hosseini R, Lamers GEM, Soltani HM, Meijer AH, Spaink HP, Schaaf MJM. Efferocytosis and extrusion of leukocytes determine the progression of early mycobacterial pathogenesis. J Cell Sci 2016; 129:3385-95. [PMID: 27469488 DOI: 10.1242/jcs.135194] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 07/25/2016] [Indexed: 12/12/2022] Open
Abstract
Macrophages and neutrophils are the first responders to invading pathogens and contribute strongly to the host defense against intracellular pathogens. The collective interplay and dynamic interactions between these leukocytes are to a large extent not understood. In the present study, we have investigated their role using a combination of confocal laser-scanning and electron microscopy in a zebrafish model for tuberculosis, a local Mycobacterium marinum infection in the tissue of the larval tail fin. Our results show that neutrophils are efficient in phagocytosis of mycobacteria and that they contribute largely to their dissemination. Macrophages appear to play a major role in efferocytosis, phagocytosis of dead cells that contain bacterial content. Phagocytic cells with large bacterial aggregates are formed that can be extruded out of the tissue after cell death. Alternatively, these excessively infected cells can undergo necrosis leading to immediate recruitment of surrounding leukocytes and subsequent phagocytosis of released bacteria. Our data show that these necrotic burst events result in progression of the infection, whereas extrusion abates the infection.
Collapse
Affiliation(s)
- Rohola Hosseini
- Institute of Biology Leiden, Leiden University, Leiden 2333CC, The Netherlands
| | - Gerda E M Lamers
- Institute of Biology Leiden, Leiden University, Leiden 2333CC, The Netherlands
| | - Hiwa M Soltani
- Institute of Biology Leiden, Leiden University, Leiden 2333CC, The Netherlands
| | - Annemarie H Meijer
- Institute of Biology Leiden, Leiden University, Leiden 2333CC, The Netherlands
| | - Herman P Spaink
- Institute of Biology Leiden, Leiden University, Leiden 2333CC, The Netherlands
| | - Marcel J M Schaaf
- Institute of Biology Leiden, Leiden University, Leiden 2333CC, The Netherlands
| |
Collapse
|
21
|
Schiebler M, Brown K, Hegyi K, Newton SM, Renna M, Hepburn L, Klapholz C, Coulter S, Obregón-Henao A, Henao Tamayo M, Basaraba R, Kampmann B, Henry KM, Burgon J, Renshaw SA, Fleming A, Kay RR, Anderson KE, Hawkins PT, Ordway DJ, Rubinsztein DC, Floto RA. Functional drug screening reveals anticonvulsants as enhancers of mTOR-independent autophagic killing of Mycobacterium tuberculosis through inositol depletion. EMBO Mol Med 2015; 7:127-39. [PMID: 25535254 PMCID: PMC4328644 DOI: 10.15252/emmm.201404137] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Mycobacterium tuberculosis (MTB) remains a major challenge to global health made worse by the spread of multidrug resistance. We therefore examined whether stimulating intracellular killing of mycobacteria through pharmacological enhancement of macroautophagy might provide a novel therapeutic strategy. Despite the resistance of MTB to killing by basal autophagy, cell-based screening of FDA-approved drugs revealed two anticonvulsants, carbamazepine and valproic acid, that were able to stimulate autophagic killing of intracellular M. tuberculosis within primary human macrophages at concentrations achievable in humans. Using a zebrafish model, we show that carbamazepine can stimulate autophagy in vivo and enhance clearance of M. marinum, while in mice infected with a highly virulent multidrug-resistant MTB strain, carbamazepine treatment reduced bacterial burden, improved lung pathology and stimulated adaptive immunity. We show that carbamazepine induces antimicrobial autophagy through a novel, evolutionarily conserved, mTOR-independent pathway controlled by cellular depletion of myo-inositol. While strain-specific differences in susceptibility to in vivo carbamazepine treatment may exist, autophagy enhancement by repurposed drugs provides an easily implementable potential therapy for the treatment of multidrug-resistant mycobacterial infection.
Collapse
Affiliation(s)
- Mark Schiebler
- Department of Medicine, Cambridge Institute for Medical Research University of Cambridge, Cambridge, UK
| | - Karen Brown
- Department of Medicine, Cambridge Institute for Medical Research University of Cambridge, Cambridge, UK Cambridge Centre for Lung Infection, Papworth Hospital, Cambridge, UK
| | - Krisztina Hegyi
- Department of Medicine, Cambridge Institute for Medical Research University of Cambridge, Cambridge, UK
| | - Sandra M Newton
- Department of Paediatric Infectious Diseases and Allergy, Imperial College London, London, UK
| | - Maurizio Renna
- Department of Medical Genetics, Cambridge Institute for Medical Research University of Cambridge, Cambridge, UK
| | - Lucy Hepburn
- Department of Medicine, Cambridge Institute for Medical Research University of Cambridge, Cambridge, UK
| | - Catherine Klapholz
- Department of Medicine, Cambridge Institute for Medical Research University of Cambridge, Cambridge, UK
| | - Sarah Coulter
- Department of Medicine, Cambridge Institute for Medical Research University of Cambridge, Cambridge, UK
| | - Andres Obregón-Henao
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Marcela Henao Tamayo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Randall Basaraba
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Beate Kampmann
- Department of Paediatric Infectious Diseases and Allergy, Imperial College London, London, UK
| | - Katherine M Henry
- Department of Infection and Immunity, University of Sheffield Western Bank, Sheffield, UK
| | - Joseph Burgon
- Department of Infection and Immunity, University of Sheffield Western Bank, Sheffield, UK
| | - Stephen A Renshaw
- Department of Infection and Immunity, University of Sheffield Western Bank, Sheffield, UK
| | - Angeleen Fleming
- Department of Medical Genetics, Cambridge Institute for Medical Research University of Cambridge, Cambridge, UK
| | - Robert R Kay
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Karen E Anderson
- The Inositide Laboratory, Babraham Institute Babraham Research Campus, Cambridge, UK
| | - Phillip T Hawkins
- The Inositide Laboratory, Babraham Institute Babraham Research Campus, Cambridge, UK
| | - Diane J Ordway
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research University of Cambridge, Cambridge, UK
| | - Rodrigo Andres Floto
- Department of Medicine, Cambridge Institute for Medical Research University of Cambridge, Cambridge, UK Cambridge Centre for Lung Infection, Papworth Hospital, Cambridge, UK
| |
Collapse
|
22
|
Sanders JL, Moulton H, Moulton Z, McLeod R, Dubey JP, Weiss LM, Zhou Y, Kent ML. The zebrafish, Danio rerio, as a model for Toxoplasma gondii: an initial description of infection in fish. JOURNAL OF FISH DISEASES 2015; 38:675-9. [PMID: 25951508 PMCID: PMC4548885 DOI: 10.1111/jfd.12393] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/23/2015] [Accepted: 04/23/2015] [Indexed: 05/04/2023]
Abstract
Toxoplasma gondii infects a very wide range of mammals and birds, and about one-third of humans are infected with this protozoan parasite. Chronic T. gondii infection has historically been believed to be asymptomatic; however there is now evidence that links chronic infection with several psychiatric disorders. While there are drugs to treat acute toxoplasmosis, there are currently no treatments for the latent form of the parasite. Currently, T. gondii in vivo research is performed using murine models, which are limited by cost and the inability to perform high throughput assays. To develop an improved in vivo model, we adapted zebrafish to 37°C and injected them intraperitoneally with two strains of T. gondii at a concentration of 10 tissue cysts per fish, and observed them for 7 days post injection. Fish were examined by histology for the presence of T. gondii development. Intracellular parasites were observed in fish at 5 to 7 days post injection. The pattern of infection observed was similar to that found in mammalian infection, with parasites developing in the somatic muscle, heart, liver, spleen, kidney, and brain.
Collapse
Affiliation(s)
- Justin L. Sanders
- Department of Microbiology, Oregon State University, Corvallis, OR
- Author for correspondence ()
| | - Hong Moulton
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR
| | | | - Rima McLeod
- Department of Ophthalmology and Visual Science, University of Chicago, Chicago, IL
| | - J. P. Dubey
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, USDA, Beltsville, MD
| | - Louis M. Weiss
- Departments of Pathology and Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Ying Zhou
- Department of Ophthalmology and Visual Science, University of Chicago, Chicago, IL
| | - Michael L. Kent
- Department of Microbiology, Oregon State University, Corvallis, OR
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR
| |
Collapse
|
23
|
Abstract
Zebrafish (Danio rerio) embryos have proven to be a powerful model for studying a variety of developmental and disease processes. External development and optical transparency make these embryos especially amenable to microscopy, and numerous transgenic lines that label specific cell types with fluorescent proteins are available, making the zebrafish embryo an ideal system for visualizing the interaction of vascular, hematopoietic, and other cell types during injury and repair in vivo. Forward and reverse genetics in zebrafish are well developed, and pharmacological manipulation is possible. We describe a mechanical vascular injury model using micromanipulation techniques that exploits several of these features to study responses to vascular injury including hemostasis and blood vessel repair. Using a combination of video and timelapse microscopy, we demonstrate that this method of vascular injury results in measurable and reproducible responses during hemostasis and wound repair. This method provides a system for studying vascular injury and repair in detail in a whole animal model.
Collapse
Affiliation(s)
- Hilary Clay
- Cardiovascular Research Institute, University of California
| | | |
Collapse
|
24
|
van Leeuwen LM, van der Sar AM, Bitter W. Animal models of tuberculosis: zebrafish. Cold Spring Harb Perspect Med 2014; 5:a018580. [PMID: 25414379 DOI: 10.1101/cshperspect.a018580] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Over the past decade the zebrafish (Danio rerio) has become an attractive new vertebrate model organism for studying mycobacterial pathogenesis. The combination of medium-throughput screening and real-time in vivo visualization has allowed new ways to dissect host pathogenic interaction in a vertebrate host. Furthermore, genetic screens on the host and bacterial sides have elucidated new mechanisms involved in the initiation of granuloma formation and the importance of a balanced immune response for control of mycobacterial pathogens. This article will highlight the unique features of the zebrafish-Mycobacterium marinum infection model and its added value for tuberculosis research.
Collapse
Affiliation(s)
- Lisanne M van Leeuwen
- Department of Pediatric Infectious Diseases and Immunology, VU University Medical Center, 1081 HV Amsterdam, The Netherlands Department of Medical Microbiology and Infection control, VU University Medical Center, 1081 BT Amsterdam, The Netherlands
| | - Astrid M van der Sar
- Department of Pediatric Infectious Diseases and Immunology, VU University Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Wilbert Bitter
- Department of Pediatric Infectious Diseases and Immunology, VU University Medical Center, 1081 HV Amsterdam, The Netherlands Department of Molecular Microbiology, VU University, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
25
|
Rowe HM, Withey JH, Neely MN. Zebrafish as a model for zoonotic aquatic pathogens. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 46:96-107. [PMID: 24607289 PMCID: PMC4096445 DOI: 10.1016/j.dci.2014.02.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/22/2014] [Accepted: 02/22/2014] [Indexed: 06/03/2023]
Abstract
Aquatic habitats harbor a multitude of bacterial species. Many of these bacteria can act as pathogens to aquatic species and/or non-aquatic organisms, including humans, that come into contact with contaminated water sources or colonized aquatic organisms. In many instances, the bacteria are not pathogenic to the aquatic species they colonize and are only considered pathogens when they come into contact with humans. There is a general lack of knowledge about how the environmental lifestyle of these pathogens allows them to persist, replicate and produce the necessary pathogenic mechanisms to successfully transmit to the human host and cause disease. Recently, the zebrafish infectious disease model has emerged as an ideal system for examining aquatic pathogens, both in the aquatic environment and during infection of the human host. This review will focus on how the zebrafish has been used successfully to analyze the pathogenesis of aquatic bacterial pathogens.
Collapse
Affiliation(s)
- Hannah M Rowe
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Jeffrey H Withey
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Melody N Neely
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
26
|
Fenaroli F, Westmoreland D, Benjaminsen J, Kolstad T, Skjeldal FM, Meijer AH, van der Vaart M, Ulanova L, Roos N, Nyström B, Hildahl J, Griffiths G. Nanoparticles as drug delivery system against tuberculosis in zebrafish embryos: direct visualization and treatment. ACS NANO 2014; 8:7014-7026. [PMID: 24945994 DOI: 10.1021/nn5019126] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Nanoparticles (NPs) enclosing antibiotics have provided promising therapy against Mycobacterium tuberculosis (Mtb) in different mammalian models. However, the NPs were not visualized in any of these animal studies. Here, we introduce the transparent zebrafish embryo as a system for noninvasive, simultaneous imaging of fluorescent NPs and the fish tuberculosis (TB) agent Mycobacterium marinum (Mm). The study was facilitated by the use of transgenic lines of macrophages, neutrophils, and endothelial cells expressing fluorescent markers readily visible in the live vertebrate. Intravenous injection of Mm led to phagocytosis by blood macrophages. These remained within the vasculature until 3 days postinfection where they started to extravasate and form aggregates of infected cells. Correlative light/electron microscopy revealed that these granuloma-like structures had significant access to the vasculature. Injection of NPs induced rapid uptake by both infected and uninfected macrophages, the latter being actively recruited to the site of infection, thereby providing an efficient targeting into granulomas. Rifampicin-loaded NPs significantly improved embryo survival and lowered bacterial load, as shown by quantitative fluorescence analysis. Our results argue that zebrafish embryos offer a powerful system for monitoring NPs in vivo and rationalize why NP therapy was so effective against Mtb in earlier studies; bacteria and NPs share the same cellular niche.
Collapse
Affiliation(s)
- Federico Fenaroli
- Department of Biosciences, University of Oslo , Blindernveien 31, 0371 Oslo, Norway
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Llewellyn MS, Boutin S, Hoseinifar SH, Derome N. Teleost microbiomes: the state of the art in their characterization, manipulation and importance in aquaculture and fisheries. Front Microbiol 2014; 5:207. [PMID: 24917852 PMCID: PMC4040438 DOI: 10.3389/fmicb.2014.00207] [Citation(s) in RCA: 307] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 04/18/2014] [Indexed: 01/10/2023] Open
Abstract
Indigenous microbiota play a critical role in the lives of their vertebrate hosts. In human and mouse models it is increasingly clear that innate and adaptive immunity develop in close concert with the commensal microbiome. Furthermore, several aspects of digestion and nutrient metabolism are governed by intestinal microbiota. Research on teleosts has responded relatively slowly to the introduction of massively parallel sequencing procedures in microbiomics. Nonetheless, progress has been made in biotic and gnotobiotic zebrafish models, defining a core microbiome and describing its role in development. However, microbiome research in other teleost species, especially those important from an aquaculture perspective, has been relatively slow. In this review, we examine progress in teleost microbiome research to date. We discuss teleost microbiomes in health and disease, microbiome ontogeny, prospects for successful microbiome manipulation (especially in an aquaculture setting) and attempt to identify important future research themes. We predict an explosion in research in this sector in line with the increasing global demand for fish protein, and the need to find sustainable approaches to improve aquaculture yield. The reduced cost and increasing ease of next generation sequencing technologies provides the technological backing, and the next 10 years will be an exciting time for teleost microbiome research.
Collapse
Affiliation(s)
- Martin S Llewellyn
- Département de Biologie, Institut de Biologie Intégrative et des Systèmes, Université Laval Québec, QC, Canada ; Molecular Ecology and Fisheries Genetics Laboratory, School of Biological Sciences, University of Wales Bangor, UK
| | - Sébastien Boutin
- Département de Biologie, Institut de Biologie Intégrative et des Systèmes, Université Laval Québec, QC, Canada
| | - Seyed Hossein Hoseinifar
- Department of Fisheries, Gorgan University of Agricultural Sciences and Natural Resources Gorgan, Iran
| | - Nicolas Derome
- Département de Biologie, Institut de Biologie Intégrative et des Systèmes, Université Laval Québec, QC, Canada
| |
Collapse
|
28
|
Host-pathogen interactions during Mycobacterium tuberculosis infections. Curr Top Microbiol Immunol 2014; 374:211-41. [PMID: 23881288 DOI: 10.1007/82_2013_332] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The intimate and persistent connection between Mycobacterium tuberculosis and its human host suggests that the pathogen has evolved extensive mechanisms to evade eradication by the immune system. In particular, the organism has adapted to replicate within phagocytic cells, especially macrophages, which are specialized to kill microbes. Over the past decade of M. tuberculosis research, the means to manipulate both the organism and the host has ushered in an exciting time that has uncovered some of the mechanisms of the innate macrophage-pathogen interactions that lie at the heart of M. tuberculosis pathogenesis, though many interactions likely still await discovery. In this chapter, we will delve into some of these advances, with an emphasis on the interactions that occur on the cellular level when M. tuberculosis cells encounter macrophages. In particular, we focus on two major aspects of M. tuberculosis biology regarding the proximal physical interface between the bacterium and host, namely the interactions with the phagosomal membrane as well as the distinctive mycobacterial cell wall. Importantly, some of the emerging paradigms in M. tuberculosis pathogenesis and host response represent common themes in bacterial pathogenesis, such as the role of host cell membrane perforation in intracellular survival and host response. However, the array of unique bacterial lipid mediators and their interaction with host cells highlights the unique biology of this persistent pathogen.
Collapse
|
29
|
Abstract
Mycobacteriosis, a chronic bacterial infection, has been associated with severe losses in some zebrafish facilities and low-level mortalities and unknown impacts in others. The occurrence of at least six different described species (Mycobacterium abscessus, M. chelonae, M. fortuitum, M. haemophilum, M. marinum, M. peregrinum) from zebrafish complicates diagnosis and control because each species is unique. As a generalization, mycobacteria are often considered opportunists, but M. haemophilum and M. marinum appear to be more virulent. Background genetics of zebrafish and environmental conditions influence the susceptibility of fish and progression of disease, emphasizing the importance of regular monitoring and good husbandry practices. A combined approach to diagnostics is ultimately the most informative, with histology as a first-level screen, polymerase chain reaction for rapid detection and species identification, and culture for strain differentiation. Occurrence of identical strains of Mycobacterium in both fish and biofilms in zebrafish systems suggests transmission can occur when fish feed on infected tissues or tank detritus containing mycobacteria. Within a facility, good husbandry practices and sentinel programs are essential for minimizing the impacts of mycobacteria. In addition, quarantine and screening of animals coming into a facility is important for eliminating the introduction of the more severe pathogens. Elimination of mycobacteria from an aquatic system is likely not feasible because these species readily establish biofilms on surfaces even in extremely low nutrient conditions. Risks associated with each commonly encountered species need to be identified and informed management plans developed. Basic research on the growth characteristics, disinfection, and pathogenesis of zebrafish mycobacteria is critical moving forward.
Collapse
Affiliation(s)
- Christopher M Whipps
- Department of Environmental and Forest Biology, State University of New York College of Environmental Science and Forestry, Syracuse, NY 13210, USA.
| | | | | |
Collapse
|
30
|
Diarylcoumarins inhibit mycolic acid biosynthesis and kill Mycobacterium tuberculosis by targeting FadD32. Proc Natl Acad Sci U S A 2013; 110:11565-70. [PMID: 23798446 DOI: 10.1073/pnas.1302114110] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Infection with the bacterial pathogen Mycobacterium tuberculosis imposes an enormous burden on global public health. New antibiotics are urgently needed to combat the global tuberculosis pandemic; however, the development of new small molecules is hindered by a lack of validated drug targets. Here, we describe the identification of a 4,6-diaryl-5,7-dimethyl coumarin series that kills M. tuberculosis by inhibiting fatty acid degradation protein D32 (FadD32), an enzyme that is required for biosynthesis of cell-wall mycolic acids. These substituted coumarin inhibitors directly inhibit the acyl-acyl carrier protein synthetase activity of FadD32. They effectively block bacterial replication both in vitro and in animal models of tuberculosis, validating FadD32 as a target for antibiotic development that works in the same pathway as the established antibiotic isoniazid. Targeting new steps in well-validated biosynthetic pathways in antitubercular therapy is a powerful strategy that removes much of the usual uncertainty surrounding new targets and in vivo clinical efficacy, while circumventing existing resistance to established targets.
Collapse
|
31
|
Wang H, Maeda Y, Fukutomi Y, Makino M. An in vitro model of Mycobacterium leprae induced granuloma formation. BMC Infect Dis 2013; 13:279. [PMID: 23782413 PMCID: PMC3693892 DOI: 10.1186/1471-2334-13-279] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 06/17/2013] [Indexed: 11/10/2022] Open
Abstract
Background Leprosy is a contagious and chronic systemic granulomatous disease caused by Mycobacterium leprae. In the pathogenesis of leprosy, granulomas play a key role, however, the mechanisms of the formation and maintenance of M. leprae granulomas are still not clearly understood. Methods To better understand the molecular physiology of M. leprae granulomas and the interaction between the bacilli and human host cells, we developed an in vitro model of human granulomas, which mimicked the in vivo granulomas of leprosy. Macrophages were differentiated from human monocytes, and infected with M. leprae, and then cultured with autologous human peripheral blood mononuclear cells (PBMCs). Results Robust granuloma-like aggregates were obtained only when the M. leprae infected macrophages were co-cultured with PBMCs. Histological examination showed M. leprae within the cytoplasmic center of the multinucleated giant cells, and these bacilli were metabolically active. Macrophages of both M1 and M2 types co-existed in the granuloma like aggregates. There was a strong relationship between the formation of granulomas and changes in the expression levels of cell surface antigens on macrophages, cytokine production and the macrophage polarization. The viability of M. leprae isolated from granulomas indicated that the formation of host cell aggregates benefited the host, but the bacilli also remained metabolically active. Conclusions A simple in vitro model of human M. leprae granulomas was established using human monocyte-derived macrophages and PBMCs. This system may be useful to unravel the mechanisms of disease progression, and subsequently develop methods to control leprosy.
Collapse
Affiliation(s)
- Hongsheng Wang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 12 Jiangwangmiao Road, Nanjing 210042, China
| | | | | | | |
Collapse
|
32
|
van der Vaart M, van Soest JJ, Spaink HP, Meijer AH. Functional analysis of a zebrafish myd88 mutant identifies key transcriptional components of the innate immune system. Dis Model Mech 2013; 6:841-54. [PMID: 23471913 PMCID: PMC3634667 DOI: 10.1242/dmm.010843] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Accepted: 01/09/2013] [Indexed: 12/17/2022] Open
Abstract
Toll-like receptors (TLRs) are an important class of pattern recognition receptors (PRRs) that recognize microbial and danger signals. Their downstream signaling upon ligand binding is vital for initiation of the innate immune response. In human and mammalian models, myeloid differentiation factor 88 (MYD88) is known for its central role as an adaptor molecule in interleukin 1 receptor (IL-1R) and TLR signaling. The zebrafish is increasingly used as a complementary model system for disease research and drug screening. Here, we describe a zebrafish line with a truncated version of MyD88 as the first zebrafish mutant for a TLR signaling component. We show that this immune-compromised mutant has a lower survival rate under standard rearing conditions and is more susceptible to challenge with the acute bacterial pathogens Edwardsiella tarda and Salmonella typhimurium. Microarray and quantitative PCR analysis revealed that expression of genes for transcription factors central to innate immunity (including NF-ĸB and AP-1) and the pro-inflammatory cytokine Il1b, is dependent on MyD88 signaling during these bacterial infections. Nevertheless, expression of immune genes independent of MyD88 in the myd88 mutant line was sufficient to limit growth of an attenuated S. typhimurium strain. In the case of infection with the chronic bacterial pathogen Mycobacterium marinum, we show that MyD88 signaling has an important protective role during early pathogenesis. During mycobacterial infection, the myd88 mutant shows accelerated formation of granuloma-like aggregates and increased bacterial burden, with associated lower induction of genes central to innate immunity. This zebrafish myd88 mutant will be a valuable tool for further study of the role of IL1R and TLR signaling in the innate immunity processes underlying infectious diseases, inflammatory disorders and cancer.
Collapse
Affiliation(s)
- Michiel van der Vaart
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Joost J. van Soest
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Herman P. Spaink
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Annemarie H. Meijer
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| |
Collapse
|
33
|
Modeling innate immune response to early Mycobacterium infection. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2012; 2012:790482. [PMID: 23365620 PMCID: PMC3529460 DOI: 10.1155/2012/790482] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 09/24/2012] [Accepted: 10/08/2012] [Indexed: 02/01/2023]
Abstract
In the study of complex patterns in biology, mathematical and computational models are emerging as important tools. In addition to experimental approaches, these modeling tools have recently been applied to address open questions regarding host-pathogen interaction dynamics, including the immune response to mycobacterial infection and tuberculous granuloma formation. We present an approach in which a computational model represents the interaction of the Mycobacterium infection with the innate immune system in zebrafish at a high level of abstraction. We use the Petri Net formalism to model the interaction between the key host elements involved in granuloma formation and infection dissemination. We define a qualitative model for the understanding and description of causal relations in this dynamic process. Complex processes involving cell-cell or cell-bacteria communication can be modeled at smaller scales and incorporated hierarchically into this main model; these are to be included in later elaborations. With the infection mechanism being defined on a higher level, lower-level processes influencing the host-pathogen interaction can be identified, modeled, and tested both quantitatively and qualitatively. This systems biology framework incorporates modeling to generate and test hypotheses, to perform virtual experiments, and to make experimentally verifiable predictions. Thereby it supports the unraveling of the mechanisms of tuberculosis infection.
Collapse
|
34
|
Li YJ, Hu B. Establishment of Multi-Site Infection Model in Zebrafish Larvae for Studying Staphylococcus aureus Infectious Disease. J Genet Genomics 2012; 39:521-34. [DOI: 10.1016/j.jgg.2012.07.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 06/26/2012] [Accepted: 07/16/2012] [Indexed: 11/30/2022]
|
35
|
Parikka M, Hammarén MM, Harjula SKE, Halfpenny NJA, Oksanen KE, Lahtinen MJ, Pajula ET, Iivanainen A, Pesu M, Rämet M. Mycobacterium marinum causes a latent infection that can be reactivated by gamma irradiation in adult zebrafish. PLoS Pathog 2012; 8:e1002944. [PMID: 23028333 PMCID: PMC3459992 DOI: 10.1371/journal.ppat.1002944] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 08/18/2012] [Indexed: 01/04/2023] Open
Abstract
The mechanisms leading to latency and reactivation of human tuberculosis are still unclear, mainly due to the lack of standardized animal models for latent mycobacterial infection. In this longitudinal study of the progression of a mycobacterial disease in adult zebrafish, we show that an experimental intraperitoneal infection with a low dose (≈ 35 bacteria) of Mycobacterium marinum, results in the development of a latent disease in most individuals. The infection is characterized by limited mortality (25%), stable bacterial loads 4 weeks following infection and constant numbers of highly organized granulomas in few target organs. The majority of bacteria are dormant during a latent mycobacterial infection in zebrafish, and can be activated by resuscitation promoting factor ex vivo. In 5-10% of tuberculosis cases in humans, the disease is reactivated usually as a consequence of immune suppression. In our model, we are able to show that reactivation can be efficiently induced in infected zebrafish by γ-irradiation that transiently depletes granulo/monocyte and lymphocyte pools, as determined by flow cytometry. This immunosuppression causes reactivation of the dormant mycobacterial population and a rapid outgrowth of bacteria, leading to 88% mortality in four weeks. In this study, the adult zebrafish presents itself as a unique non-mammalian vertebrate model for studying the development of latency, regulation of mycobacterial dormancy, as well as reactivation of latent or subclinical tuberculosis. The possibilities for screening for host and pathogen factors affecting the disease progression, and identifying novel therapeutic agents and vaccine targets make this established model especially attractive.
Collapse
|
36
|
Brothers KM, Wheeler RT. Non-invasive imaging of disseminated candidiasis in zebrafish larvae. J Vis Exp 2012:4051. [PMID: 22872032 DOI: 10.3791/4051] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Disseminated candidiasis caused by the pathogen Candida albicans is a clinically important problem in hospitalized individuals and is associated with a 30 to 40% attributable mortality(6). Systemic candidiasis is normally controlled by innate immunity, and individuals with genetic defects in innate immune cell components such as phagocyte NADPH oxidase are more susceptible to candidemia(7-9). Very little is known about the dynamics of C. albicans interaction with innate immune cells in vivo. Extensive in vitro studies have established that outside of the host C. albicans germinates inside of macrophages, and is quickly destroyed by neutrophils(10-14). In vitro studies, though useful, cannot recapitulate the complex in vivo environment, which includes time-dependent dynamics of cytokine levels, extracellular matrix attachments, and intercellular contacts(10, 15-18). To probe the contribution of these factors in host-pathogen interaction, it is critical to find a model organism to visualize these aspects of infection non-invasively in a live intact host. The zebrafish larva offers a unique and versatile vertebrate host for the study of infection. For the first 30 days of development zebrafish larvae have only innate immune defenses(2, 19-21), simplifying the study of diseases such as disseminated candidiasis that are highly dependent on innate immunity. The small size and transparency of zebrafish larvae enable imaging of infection dynamics at the cellular level for both host and pathogen. Transgenic larvae with fluorescing innate immune cells can be used to identify specific cells types involved in infection(22-24). Modified anti-sense oligonucleotides (Morpholinos) can be used to knock down various immune components such as phagocyte NADPH oxidase and study the changes in response to fungal infection(5). In addition to the ethical and practical advantages of using a small lower vertebrate, the zebrafish larvae offers the unique possibility to image the pitched battle between pathogen and host both intravitally and in color. The zebrafish has been used to model infection for a number of human pathogenic bacteria, and has been instrumental in major advances in our understanding of mycobacterial infection(3, 25). However, only recently have much larger pathogens such as fungi been used to infect larva(5, 23, 26), and to date there has not been a detailed visual description of the infection methodology. Here we present our techniques for hindbrain ventricle microinjection of prim(25) zebrafish, including our modifications to previous protocols. Our findings using the larval zebrafish model for fungal infection diverge from in vitro studies and reinforce the need to examine the host-pathogen interaction in the complex environment of the host rather than the simplified system of the Petri dish(5).
Collapse
|
37
|
Rendueles O, Ferrières L, Frétaud M, Bégaud E, Herbomel P, Levraud JP, Ghigo JM. A new zebrafish model of oro-intestinal pathogen colonization reveals a key role for adhesion in protection by probiotic bacteria. PLoS Pathog 2012; 8:e1002815. [PMID: 22911651 PMCID: PMC3406073 DOI: 10.1371/journal.ppat.1002815] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 06/09/2012] [Indexed: 11/19/2022] Open
Abstract
The beneficial contribution of commensal bacteria to host health and homeostasis led to the concept that exogenous non-pathogenic bacteria called probiotics could be used to limit disease caused by pathogens. However, despite recent progress using gnotobiotic mammal and invertebrate models, mechanisms underlying protection afforded by commensal and probiotic bacteria against pathogens remain poorly understood. Here we developed a zebrafish model of controlled co-infection in which germ-free zebrafish raised on axenic living protozoa enabled the study of interactions between host and commensal and pathogenic bacteria. We screened enteric fish pathogens and identified Edwardsiella ictaluri as a virulent strain inducing a strong inflammatory response and rapid mortality in zebrafish larvae infected by the natural oro-intestinal route. Using mortality induced by infection as a phenotypic read-out, we pre-colonized zebrafish larvae with 37 potential probiotic bacterial strains and screened for survival upon E. ictaluri infection. We identified 3 robustly protective strains, including Vibrio parahaemolyticus and 2 Escherichia coli strains. We showed that the observed protective effect of E. coli was not correlated with a reduced host inflammatory response, nor with the release of biocidal molecules by protective bacteria, but rather with the presence of specific adhesion factors such as F pili that promote the emergence of probiotic bacteria in zebrafish larvae. Our study therefore provides new insights into the molecular events underlying the probiotic effect and constitutes a potentially high-throughput in vivo approach to the study of the molecular basis of pathogen exclusion in a relevant model of vertebrate oro-intestinal infection.
Collapse
Affiliation(s)
- Olaya Rendueles
- Institut Pasteur, Unité de Génétique des Biofilms, Département de Microbiologie, Paris, France
| | - Lionel Ferrières
- Institut Pasteur, Unité de Génétique des Biofilms, Département de Microbiologie, Paris, France
| | - Maxence Frétaud
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Département de Biologie du Développement, Paris, France
- CNRS, URA2578, Paris, France
| | - Evelyne Bégaud
- Institut Pasteur, Centre de Ressources Biologiques de l'Institut Pasteur, Paris, France
| | - Philippe Herbomel
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Département de Biologie du Développement, Paris, France
- CNRS, URA2578, Paris, France
| | - Jean-Pierre Levraud
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Département de Biologie du Développement, Paris, France
- CNRS, URA2578, Paris, France
| | - Jean-Marc Ghigo
- Institut Pasteur, Unité de Génétique des Biofilms, Département de Microbiologie, Paris, France
- * E-mail:
| |
Collapse
|
38
|
Renshaw SA, Trede NS. A model 450 million years in the making: zebrafish and vertebrate immunity. Dis Model Mech 2012; 5:38-47. [PMID: 22228790 PMCID: PMC3255542 DOI: 10.1242/dmm.007138] [Citation(s) in RCA: 253] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Since its first splash 30 years ago, the use of the zebrafish model has been extended from a tool for genetic dissection of early vertebrate development to the functional interrogation of organogenesis and disease processes such as infection and cancer. In particular, there is recent and growing attention in the scientific community directed at the immune systems of zebrafish. This development is based on the ability to image cell movements and organogenesis in an entire vertebrate organism, complemented by increasing recognition that zebrafish and vertebrate immunity have many aspects in common. Here, we review zebrafish immunity with a particular focus on recent studies that exploit the unique genetic and in vivo imaging advantages available for this organism. These unique advantages are driving forward our study of vertebrate immunity in general, with important consequences for the understanding of mammalian immune function and its role in disease pathogenesis.
Collapse
Affiliation(s)
- Stephen A Renshaw
- MRC Centre for Developmental and Biomedical Genetics, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| | | |
Collapse
|
39
|
Hess I, Boehm T. Intravital imaging of thymopoiesis reveals dynamic lympho-epithelial interactions. Immunity 2012; 36:298-309. [PMID: 22342843 DOI: 10.1016/j.immuni.2011.12.016] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 11/24/2011] [Accepted: 12/22/2011] [Indexed: 12/17/2022]
Abstract
T cell development occurs in the thymus. The thymic microenvironment attracts hematopoietic progenitors, specifies them toward the T cell lineage, and orchestrates their differentiation and egress into the periphery. The anatomical location of the thymus and the intrauterine development of mouse embryos have so far precluded a direct visualization of the initial steps of thymopoiesis. Here, we describe transgenic zebrafish lines enabling the in vivo observation of thymopoiesis. The cell-autonomous proliferation of thymic epithelial cells, their morphological transformation into a reticular meshwork upon contact with hematopoietic cells, and the multiple migration routes of thymus-settling cells could be directly visualized. The unexpectedly dynamic thymus homing process is chemokine driven and independent of blood circulation. Thymocyte development appears to be completed in less than 4 days. Our work establishes a versatile model for the in vivo observation and manipulation of thymopoiesis.
Collapse
Affiliation(s)
- Isabell Hess
- Department of Developmental Immunology, Max-Planck Institute of Immunobiology and Epigenetics, Stuebeweg 51, 79108 Freiburg, Germany
| | | |
Collapse
|
40
|
van der Woude AD, Sarkar D, Bhatt A, Sparrius M, Raadsen SA, Boon L, Geurtsen J, van der Sar AM, Luirink J, Houben ENG, Besra GS, Bitter W. Unexpected link between lipooligosaccharide biosynthesis and surface protein release in Mycobacterium marinum. J Biol Chem 2012; 287:20417-29. [PMID: 22505711 DOI: 10.1074/jbc.m111.336461] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The mycobacterial cell envelope is characterized by the presence of a highly impermeable second membrane, which is composed of mycolic acids intercalated with different unusual free lipids, such as lipooligosaccharides (LOS). Transport across this cell envelope requires a dedicated secretion system for extracellular proteins, such as PE_PGRS proteins, which are specific mycobacterial proteins with polymorphic GC-rich sequence (PGRS). In this study, we set out to identify novel components involved in the secretion of PE_PGRS proteins by screening Mycobacterium marinum transposon mutants for secretion defects. Interestingly, most mutants were not affected in secretion but in the release of PE_PGRS proteins from the cell surface. These mutants had insertions in a gene cluster associated with LOS biosynthesis. Lipid analysis of these mutants revealed a role at different stages of LOS biosynthesis for 10 novel genes. Furthermore, we show that regulatory protein WhiB4 is involved in LOS biosynthesis. The absence of the most extended LOS molecule, i.e. LOS-IV, and a concomitant accumulation of LOS-III was already sufficient to reduce the release of PE_PGRS proteins from the mycobacterial cell surface. A similar effect was observed for major surface protein EspE. These results show that the attachment of surface proteins is strongly influenced by the glycolipid composition of the mycobacterial cell envelope. Finally, we tested the virulence of a LOS-IV-deficient mutant in our zebrafish embryo infection model. This mutant showed a marked increase in virulence as compared with the wild-type strain, suggesting that LOS-IV plays a role in the modulation of mycobacterial virulence.
Collapse
Affiliation(s)
- Aniek D van der Woude
- Department of Medical Microbiology and Infection Control, VU University Medical Center, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Grayfer L, Hodgkinson JW, Belosevic M. Analysis of the antimicrobial responses of primary phagocytes of the goldfish (Carassius auratus L.) against Mycobacterium marinum. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:1146-1158. [PMID: 21530582 DOI: 10.1016/j.dci.2011.04.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 04/11/2011] [Accepted: 04/12/2011] [Indexed: 05/30/2023]
Abstract
The slow growth rate of Mycobacterium spp. that infect humans coupled with a lack of reliable in vitro infection model systems has hindered the progress of research in host cell-mycobacteria interactions. Recent studies have utilized the relatively fast growing Mycobacterium marinum to examine the host-pathogen interface in natural fish hosts. Here we describe the use of primary goldfish monocyte and mature macrophage cultures to investigate the immune cell-M. marinum interactions. Live and heat-killed M. marinum abrogated the recombinant goldfish (rg)TNFα2 and rgIFNγ-induced monocyte reactive oxygen production. Live but not heat-killed M. marinum also ablated rgIFNγrel and rg-TNFα2 induced macrophage nitric oxide production. M. marinum induced significant changes in gene expression of select NADPH oxidase components and inflammatory cytokine receptors and up-regulated the expression of immunosuppressive genes IL-10, TGFβ1 and SOCS-3. The exposure of monocytes and mature macrophages to M. marinum caused an increase in the mRNA levels of several pro-inflammatory genes. Stimulation of monocytes and macrophages with rgTNFα2, rgIFNγ, or rgIFNγrel reduced the survival of intracellular mycobacteria. The characterization of the interaction between M. marinum and natural host-derived primary phagocyte cultures will enable future studies on the host-pathogen interactions in mycobacterial infections.
Collapse
Affiliation(s)
- Leon Grayfer
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
42
|
Meijer AH, Spaink HP. Host-pathogen interactions made transparent with the zebrafish model. Curr Drug Targets 2011; 12:1000-17. [PMID: 21366518 PMCID: PMC3319919 DOI: 10.2174/138945011795677809] [Citation(s) in RCA: 176] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 10/21/2010] [Indexed: 01/18/2023]
Abstract
The zebrafish holds much promise as a high-throughput drug screening model for immune-related diseases, including inflammatory and infectious diseases and cancer. This is due to the excellent possibilities for in vivo imaging in combination with advanced tools for genomic and large scale mutant analysis. The context of the embryo’s developing immune system makes it possible to study the contribution of different immune cell types to disease progression. Furthermore, due to the temporal separation of innate immunity from adaptive responses, zebrafish embryos and larvae are particularly useful for dissecting the innate host factors involved in pathology. Recent studies have underscored the remarkable similarity of the zebrafish and human immune systems, which is important for biomedical applications. This review is focused on the use of zebrafish as a model for infectious diseases, with emphasis on bacterial pathogens. Following a brief overview of the zebrafish immune system and the tools and methods used to study host-pathogen interactions in zebrafish, we discuss the current knowledge on receptors and downstream signaling components that are involved in the zebrafish embryo’s innate immune response. We summarize recent insights gained from the use of bacterial infection models, particularly the Mycobacterium marinum model, that illustrate the potential of the zebrafish model for high-throughput antimicrobial drug screening.
Collapse
Affiliation(s)
- Annemarie H Meijer
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| | | |
Collapse
|
43
|
Shiryaev SA, Cieplak P, Aleshin AE, Sun Q, Zhu W, Motamedchaboki K, Sloutsky A, Strongin AY. Matrix metalloproteinase proteolysis of the mycobacterial HSP65 protein as a potential source of immunogenic peptides in human tuberculosis. FEBS J 2011; 278:3277-86. [PMID: 21752195 DOI: 10.1111/j.1742-4658.2011.08244.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mycobacterium tuberculosis is the causative agent of human tuberculosis (TB). Mycobacterial secretory protein ESAT-6 induces matrix metalloproteinase (MMP)-9 in epithelial cells neighboring infected macrophages. MMP-9 then enhances recruitment of uninfected macrophages, which contribute to nascent granuloma maturation and bacterial growth. Disruption of MMP-9 function attenuates granuloma formation and bacterial growth. The abundant mycobacterial 65 kDa heat shock protein (HSP65) chaperone is the major target for the immune response and a critical component in M. tuberculosis adhesion to macrophages. We hypothesized that HSP65 is susceptible to MMP-9 proteolysis and that the resulting HSP65 immunogenic peptides affect host adaptive immunity. To identify MMPs that cleave HSP65, we used MMP-2 and MMP-9 gelatinases, the simple hemopexin domain MMP-8, membrane-associated MMP-14, MMP-15, MMP-16 and MMP-24, and glycosylphosphatidylinositol-linked MMP-17 and MMP-25. We determined both the relative cleavage efficiency of MMPs against the HSP65 substrate and the peptide sequence of the cleavage sites. Cleavage of the unstructured PAGHG474L C-terminal region initiates the degradation of HSP65 by MMPs. This initial cleavage destroys the substrate-binding capacity of the HSP65 chaperone. Multiple additional cleavages of the unfolded HSP65 then follow. MMP-2, MMP-8, MMP-14, MMP-15 and MMP-16, in addition to MMP-9, generate the known highly immunogenic N-terminal peptide of HSP65. Based on our biochemical data, we now suspect that MMP proteolysis of HSP65 in vivo, including MMP-9 proteolysis, also results in the abundant generation of the N-terminal immunogenic peptide and that this peptide, in addition to intact HSP65, contributes to the complex immunomodulatory interplay in the course of TB infection.
Collapse
Affiliation(s)
- Sergey A Shiryaev
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Live imaging of disseminated candidiasis in zebrafish reveals role of phagocyte oxidase in limiting filamentous growth. EUKARYOTIC CELL 2011; 10:932-44. [PMID: 21551247 DOI: 10.1128/ec.05005-11] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Candida albicans is a human commensal and a clinically important fungal pathogen that grows in both yeast and hyphal forms during human infection. Although Candida can cause cutaneous and mucosal disease, systemic infections cause the greatest mortality in hospitals. Candidemia occurs primarily in immunocompromised patients, for whom the innate immune system plays a paramount role in immunity. We have developed a novel transparent vertebrate model of candidemia to probe the molecular nature of Candida-innate immune system interactions in an intact host. Our zebrafish infection model results in a lethal disseminated disease that shares important traits with disseminated candidiasis in mammals, including dimorphic fungal growth, dependence on hyphal growth for virulence, and dependence on the phagocyte NADPH oxidase for immunity. Dual imaging of fluorescently marked immune cells and fungi revealed that phagocytosed yeast cells can remain viable and even divide within macrophages without germinating. Similarly, although we observed apparently killed yeast cells within neutrophils, most yeast cells within these innate immune cells were viable. Exploiting this model, we combined intravital imaging with gene knockdown to show for the first time that NADPH oxidase is required for regulation of C. albicans filamentation in vivo. The transparent and easily manipulated larval zebrafish model promises to provide a unique tool for dissecting the molecular basis of phagocyte NADPH oxidase-mediated limitation of filamentous growth in vivo.
Collapse
|
45
|
Carvalho R, de Sonneville J, Stockhammer OW, Savage NDL, Veneman WJ, Ottenhoff THM, Dirks RP, Meijer AH, Spaink HP. A high-throughput screen for tuberculosis progression. PLoS One 2011; 6:e16779. [PMID: 21390204 PMCID: PMC3040195 DOI: 10.1371/journal.pone.0016779] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Accepted: 12/29/2010] [Indexed: 12/16/2022] Open
Abstract
One-third of the world population is infected with Mycobacterium tuberculosis and multi-drug resistant strains are rapidly evolving. The noticeable absence of a whole organism high-throughput screening system for studying the progression of tuberculosis is fast becoming the bottleneck in tuberculosis research. We successfully developed such a system using the zebrafish Mycobacterium marinum infection model, which is a well-characterized model for tuberculosis progression with biomedical significance, mimicking hallmarks of human tuberculosis pathology. Importantly, we demonstrate the suitability of our system to directly study M. tuberculosis, showing for the first time that the human pathogen can propagate in this vertebrate model, resulting in similar early disease symptoms to those observed upon M. marinum infection. Our system is capable of screening for disease progression via robotic yolk injection of early embryos and visual flow screening of late-stage larvae. We also show that this system can reliably recapitulate the standard caudal vein injection method with a throughput level of 2,000 embryos per hour. We additionally demonstrate the possibility of studying signal transduction leading to disease progression using reverse genetics at high-throughput levels. Importantly, we use reference compounds to validate our system in the testing of molecules that prevent tuberculosis progression, making it highly suited for investigating novel anti-tuberculosis compounds in vivo.
Collapse
|
46
|
Castaño D, Barrera LF, Rojas M. Mycobacterium tuberculosis alters the differentiation of monocytes into macrophages in vitro. Cell Immunol 2011; 268:60-7. [DOI: 10.1016/j.cellimm.2011.02.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 02/22/2011] [Accepted: 02/23/2011] [Indexed: 01/21/2023]
|
47
|
Cui C, Benard EL, Kanwal Z, Stockhammer OW, van der Vaart M, Zakrzewska A, Spaink HP, Meijer AH. Infectious disease modeling and innate immune function in zebrafish embryos. Methods Cell Biol 2011; 105:273-308. [PMID: 21951535 DOI: 10.1016/b978-0-12-381320-6.00012-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The major cell types of the innate immune system, macrophages and neutrophils, develop during the first two days of zebrafish embryogenesis. The interaction of these immune cells with pathogenic microbes can excellently be traced in the optically transparent zebrafish embryos. Various tools and methods have recently been developed for visualizing and isolating the zebrafish embryonic innate immune cells, for establishing infections by different micro-injection techniques, and for analyzing the host innate immune response following microbial recognition. Here we provide practical guidelines for the application of these methodologies and review the current state of the art in zebrafish infectious disease research.
Collapse
Affiliation(s)
- Chao Cui
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Curtis J, Kopanitsa L, Stebbings E, Speirs A, Ignatyeva O, Balabanova Y, Nikolayevskyy V, Hoffner S, Horstmann R, Drobniewski F, Nejentsev S. Association analysis of the LTA4H gene polymorphisms and pulmonary tuberculosis in 9115 subjects. Tuberculosis (Edinb) 2010; 91:22-5. [PMID: 21112816 PMCID: PMC3040790 DOI: 10.1016/j.tube.2010.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2010] [Revised: 09/20/2010] [Accepted: 11/07/2010] [Indexed: 01/04/2023]
Abstract
Immunoregulatory eicosanoids have been implicated in protection from mycobacterial infection in cell and animal models. Recently, a study of the zebrafish embryo demonstrated that mutants of the lta4h gene, which encodes the leukotriene A4 hydrolase (LTA4H) enzyme of the eicosanoid pathway, have hypersusceptibility to Mycobacterium marinum infection. It also reported that heterozygosity at the two single nucleotide polymorphisms rs1978331 and rs2660898 located in introns of the LTA4H gene, a human homologue of lta4h, is associated with protection from pulmonary tuberculosis. To replicate this association we genotyped six LTA4H gene polymorphisms in samples from 3703 pulmonary tuberculosis patients and 5412 healthy controls collected in Russia. We found no evidence of the protective effect of heterozygosity at the polymorphisms rs1978331 and rs2660898 (P = 0.29 and 0.49) and no association of the alleles of any of the six polymorphisms (P = 0.13-0.81). These results suggest that common polymorphisms in the LTA4H gene do not play any major role in susceptibility to clinical pulmonary tuberculosis.
Collapse
Affiliation(s)
- James Curtis
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom, Level 5, Addenbrookes Hospital, Hills road, Cambridge CB2 0QQ, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Stockhammer OW, Rauwerda H, Wittink FR, Breit TM, Meijer AH, Spaink HP. Transcriptome analysis of Traf6 function in the innate immune response of zebrafish embryos. Mol Immunol 2010; 48:179-90. [PMID: 20851470 DOI: 10.1016/j.molimm.2010.08.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 08/11/2010] [Accepted: 08/17/2010] [Indexed: 10/19/2022]
Abstract
TRAF6 is a key player at the cross-roads of development and immunity. The analysis of its in vivo molecular function is a great challenge since severe developmental defects and early lethality caused by Traf6 deficiency in knock-out mice interfere with analyses of the immune response. In this study we have used a new strategy to analyze the function of Traf6 in a zebrafish-Salmonella infectious disease model. In our approach the effect of a Traf6 translation-blocking morpholino was titrated down to avoid developmental defects and the response to infection under these conditions was studied using the combination of microarray analysis and whole transcriptome deep sequencing. Transcriptome profiling of the traf6 knock-down allowed the identification of a gene set whose responsiveness during infection is highly dependent on Traf6. Expression trend analysis based on the resulting datasets identified nine clusters of genes with characteristic transcription response profiles, demonstrating Traf6 has a dynamic role as a positive and negative regulator. Among the Traf6-dependent genes was a large set of well known anti-microbial and inflammatory genes. Additionally, we identified several genes which were not previously linked to a response to microbial infection, such as the fertility hormone gene gnrh2 and the DNA-damage regulated autophagy modulator 1 gene dram1. With the use of the zebrafish embryo model we have now analyzed the in vivo function of Traf6 in the innate immune response without interference of adaptive immunity.
Collapse
Affiliation(s)
- Oliver W Stockhammer
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | | | | | | | | | | |
Collapse
|
50
|
Eosinophils in the zebrafish: prospective isolation, characterization, and eosinophilia induction by helminth determinants. Blood 2010; 116:3944-54. [PMID: 20713961 DOI: 10.1182/blood-2010-03-267419] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Eosinophils are granulocytic leukocytes implicated in numerous aspects of immunity and disease. The precise functions of eosinophils, however, remain enigmatic. Alternative models to study eosinophil biology may thus yield novel insights into their function. Eosinophilic cells have been observed in zebrafish but have not been thoroughly characterized. We used a gata2:eGFP transgenic animal to enable prospective isolation and characterization of zebrafish eosinophils, and demonstrate that all gata2(hi) cells in adult hematopoietic tissues are eosinophils. Although eosinophils are rare in most organs, they are readily isolated from whole kidney marrow and abundant within the peritoneal cavity. Molecular analyses demonstrate that zebrafish eosinophils express genes important for the activities of mammalian eosinophils. In addition, gata2(hi) cells degranulate in response to helminth extract. Chronic exposure to helminth- related allergens resulted in profound eosinophilia, demonstrating that eosinophil responses to allergens have been conserved over evolution. Importantly, infection of adult zebrafish with Pseudocapillaria tomentosa, a natural nematode pathogen of teleosts, caused marked increases in eosinophil number within the intestine. Together, these observations support a conserved role for eosinophils in the response to helminth antigens or infection and provide a new model to better understand how parasitic worms activate, co-opt, or evade the vertebrate immune response.
Collapse
|