Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Nature ; 517(7536): 612-5, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25470057

ABSTRACT

Pathogenic mycobacteria induce the formation of complex cellular aggregates called granulomas that are the hallmark of tuberculosis. Here we examine the development and consequences of vascularization of the tuberculous granuloma in the zebrafish-Mycobacterium marinum infection model, which is characterized by organized granulomas with necrotic cores that bear striking resemblance to those of human tuberculosis. Using intravital microscopy in the transparent larval zebrafish, we show that granuloma formation is intimately associated with angiogenesis. The initiation of angiogenesis in turn coincides with the generation of local hypoxia and transcriptional induction of the canonical pro-angiogenic molecule Vegfaa. Pharmacological inhibition of the Vegf pathway suppresses granuloma-associated angiogenesis, reduces infection burden and limits dissemination. Moreover, anti-angiogenic therapies synergize with the first-line anti-tubercular antibiotic rifampicin, as well as with the antibiotic metronidazole, which targets hypoxic bacterial populations. Our data indicate that mycobacteria induce granuloma-associated angiogenesis, which promotes mycobacterial growth and increases spread of infection to new tissue sites. We propose the use of anti-angiogenic agents, now being used in cancer regimens, as a host-targeting tuberculosis therapy, particularly in extensively drug-resistant disease for which current antibiotic regimens are largely ineffective.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/drug effects , Mycobacterium marinum/growth & development , Neovascularization, Pathologic/microbiology , Signal Transduction/drug effects , Zebrafish/microbiology , Angiogenesis Inhibitors/therapeutic use , Animals , Antibiotics, Antitubercular/pharmacology , Bacterial Load/drug effects , Disease Models, Animal , Drug Synergism , Granuloma/drug therapy , Granuloma/metabolism , Granuloma/microbiology , Granuloma/pathology , Hypoxia/metabolism , Hypoxia/microbiology , Hypoxia/pathology , Larva/drug effects , Larva/microbiology , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/metabolism , Mycobacterium Infections, Nontuberculous/pathology , Mycobacterium marinum/pathogenicity , Neovascularization, Pathologic/drug therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/metabolism , Tuberculosis/drug therapy , Tuberculosis/microbiology , Tuberculosis/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Zebrafish/growth & development
2.
Cell Microbiol ; 21(11): e13083, 2019 11.
Article in English | MEDLINE | ID: mdl-31290267

ABSTRACT

Integrated with both a historical perspective and an evolutionary angle, this opinion article presents a brief and personal view of the emergence of cellular microbiology research. From the very first observations of phagocytosis by Goeze in 1777 to the exhaustive analysis of the cellular defence mechanisms performed in modern laboratories, the studies by cell biologists and microbiologists have converged into an integrative research field distinct from, but fully coupled to immunity: cellular microbiology. In addition, this brief article is thought as a humble patchwork of the motivations that have guided the research in my group over a quarter century.


Subject(s)
Dictyostelium/immunology , Mycobacterium marinum/immunology , Phagocytosis/immunology , Animals , Dictyostelium/growth & development , Dictyostelium/microbiology , Dictyostelium/ultrastructure , History, 18th Century , History, 19th Century , History, 21st Century , Host-Pathogen Interactions , Humans , Immunity, Innate , Microbiology/history , Mycobacterium marinum/growth & development , Mycobacterium marinum/pathogenicity , Phagosomes/immunology , Phagosomes/microbiology , Phagosomes/ultrastructure
3.
Cell Microbiol ; 21(6): e13008, 2019 06.
Article in English | MEDLINE | ID: mdl-30656819

ABSTRACT

The causative agent of tuberculosis, Mycobacterium tuberculosis, and its close relative Mycobacterium marinum manipulate phagocytic host cells, thereby creating a replication-permissive compartment termed the Mycobacterium-containing vacuole (MCV). The phosphoinositide (PI) lipid pattern is a crucial determinant of MCV formation and is targeted by mycobacterial PI phosphatases. In this study, we establish an efficient phage transduction protocol to construct defined M. marinum deletion mutants lacking one or three phosphatases, PtpA, PtpB, and/or SapM. These strains were defective for intracellular replication in macrophages and amoebae, and the growth defect was complemented by the corresponding plasmid-borne genes. Fluorescence microscopy of M. marinum-infected Dictyostelium discoideum revealed that MCVs harbouring mycobacteria lacking PtpA, SapM, or all three phosphatases accumulate significantly more phosphatidylinositol-3-phosphate (PtdIns3P) compared with MCVs containing the parental strain. Moreover, PtpA reduced MCV acidification by blocking the recruitment of the V-ATPase, and all three phosphatases promoted bacterial escape from the pathogen vacuole to the cytoplasm. In summary, the secreted M. marinum phosphatases PtpA, PtpB, and SapM determine the MCV PI pattern, compartment acidification, and phagosomal escape.


Subject(s)
Cytosol/metabolism , Mycobacterium marinum/growth & development , Phagosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/metabolism , Vacuoles/metabolism , Acanthamoeba castellanii/microbiology , Adenosine Triphosphatases/metabolism , Amoeba/microbiology , Animals , Bacterial Proteins/metabolism , Dictyostelium/metabolism , Dictyostelium/microbiology , Host-Pathogen Interactions/genetics , Macrophages/enzymology , Macrophages/microbiology , Mice , Microscopy, Fluorescence , Mycobacterium marinum/enzymology , Mycobacterium marinum/genetics , Mycobacterium marinum/pathogenicity , Protein Tyrosine Phosphatases/metabolism , RAW 264.7 Cells , Vacuoles/microbiology
4.
Cell Microbiol ; 20(9): e12858, 2018 09.
Article in English | MEDLINE | ID: mdl-29749044

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.


Subject(s)
Blood-Brain Barrier/microbiology , Central Nervous System Infections/pathology , Host-Pathogen Interactions , Mycobacterium Infections, Nontuberculous/pathology , Mycobacterium marinum/growth & development , Animals , Brain/microbiology , Disease Models, Animal , Macrophages/microbiology , Zebrafish
5.
J Antimicrob Chemother ; 72(2): 421-427, 2017 02.
Article in English | MEDLINE | ID: mdl-27798206

ABSTRACT

OBJECTIVES: Mycobacterium tuberculosis is a deadly human pathogen that causes the lung disease TB. M. tuberculosis latently infects a third of the world's population, resulting in ∼1.5 million deaths per year. Due to the difficulties and expense of carrying out animal drug trials using M. tuberculosis and rodents, infections of the zebrafish Danio rerio with Mycobacterium marinum have become a useful surrogate. However, the infection methods described to date require specialized equipment and a high level of operator expertise. METHODS: We investigated whether zebrafish larvae could be naturally infected with bioluminescently labelled M. marinum by immersion, and whether infected larvae could be used for rapid screening of anti-mycobacterial compounds using bioluminescence. We used rifampicin and a variety of nitroimidazole-based next-generation and experimental anti-mycobacterial drugs, selected for their wide range of potencies against M. tuberculosis, to validate this model for anti-mycobacterial drug discovery. RESULTS: We observed that five of the six treatments (rifampicin, pretomanid, delamanid, SN30488 and SN30527) significantly reduced the bioluminescent signal from M. marinum within naturally infected zebrafish larvae. Importantly, these same five treatments also retarded the growth of M. tuberculosis in vitro. In contrast, only three of the six treatments tested (rifampicin, delamanid and SN30527) retarded the growth of M. marinum in vitro. CONCLUSIONS: We have demonstrated that zebrafish larvae naturally infected with bioluminescent M. marinum M can be used for the rapid screening of anti-mycobacterial compounds with readily available equipment and limited expertise. The result is an assay that can be carried out by a wide variety of laboratories for minimal cost and without high levels of zebrafish expertise.


Subject(s)
Antitubercular Agents/isolation & purification , Antitubercular Agents/pharmacology , Drug Evaluation, Preclinical/methods , Mycobacterium marinum/drug effects , Zebrafish/microbiology , Animals , Larva/microbiology , Luminescent Measurements , Mycobacterium marinum/growth & development , Nitroimidazoles/pharmacology , Rifampin/pharmacology , Staining and Labeling
6.
Immunity ; 29(2): 283-94, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18691913

ABSTRACT

Tumor necrosis factor (TNF), a key effector in controlling tuberculosis, is thought to exert protection by directing formation of granulomas, organized aggregates of macrophages and other immune cells. Loss of TNF signaling causes progression of tuberculosis in humans, and the increased mortality of Mycobacterium tuberculosis-infected mice is associated with disorganized necrotic granulomas, although the precise roles of TNF signaling preceding this endpoint remain undefined. We monitored transparent Mycobacterium marinum-infected zebrafish live to conduct a stepwise dissection of how TNF signaling operates in mycobacterial pathogenesis. We found that loss of TNF signaling caused increased mortality even when only innate immunity was operant. In the absence of TNF, intracellular bacterial growth and granuloma formation were accelerated and was followed by necrotic death of overladen macrophages and granuloma breakdown. Thus, TNF is not required for tuberculous granuloma formation, but maintains granuloma integrity indirectly by restricting mycobacterial growth within macrophages and preventing their necrosis.


Subject(s)
Granuloma/immunology , Macrophages/immunology , Macrophages/microbiology , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium marinum/immunology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Tumor Necrosis Factors/metabolism , Animals , Apoptosis , Cell Death , Cell Movement , Cytokines/immunology , Cytokines/metabolism , Embryo, Nonmammalian , Granuloma/metabolism , Granuloma/microbiology , Immunity, Innate , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/growth & development , Mycobacterium marinum/physiology , Receptors, Tumor Necrosis Factor, Type I/immunology , Tumor Necrosis Factors/immunology , Zebrafish/immunology , Zebrafish/microbiology
7.
J Biol Chem ; 290(21): 13321-43, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25825498

ABSTRACT

Mycobacterium tuberculosis employs various strategies to modulate host immune responses to facilitate its persistence in macrophages. The M. tuberculosis cell wall contains numerous glycoproteins with unknown roles in pathogenesis. Here, by using Concanavalin A and LC-MS analysis, we identified a novel mannosylated glycoprotein phosphoribosyltransferase, encoded by Rv3242c from M. tuberculosis cell walls. Homology modeling, bioinformatic analyses, and an assay of phosphoribosyltransferase activity in Mycobacterium smegmatis expressing recombinant Rv3242c (MsmRv3242c) confirmed the mass spectrometry data. Using Mycobacterium marinum-zebrafish and the surrogate MsmRv3242c infection models, we proved that phosphoribosyltransferase is involved in mycobacterial virulence. Histological and infection assays showed that the M. marinum mimG mutant, an Rv3242c orthologue in a pathogenic M. marinum strain, was strongly attenuated in adult zebrafish and also survived less in macrophages. In contrast, infection with wild type and the complemented ΔmimG:Rv3242c M. marinum strains showed prominent pathological features, such as severe emaciation, skin lesions, hemorrhaging, and more zebrafish death. Similarly, recombinant MsmRv3242c bacteria showed increased invasion in non-phagocytic epithelial cells and longer intracellular survival in macrophages as compared with wild type and vector control M. smegmatis strains. Further mechanistic studies revealed that the Rv3242c- and mimG-mediated enhancement of intramacrophagic survival was due to inhibition of autophagy, reactive oxygen species, and reduced activities of superoxide dismutase and catalase enzymes. Infection with MsmRv3242c also activated the MAPK pathway, NF-κB, and inflammatory cytokines. In summary, we show that a novel mycobacterial mannosylated phosphoribosyltransferase acts as a virulence and immunomodulatory factor, suggesting that it may constitute a novel target for antimycobacterial drugs.


Subject(s)
Autophagy , Macrophages/immunology , Mycobacterium marinum/pathogenicity , Mycobacterium tuberculosis/pathogenicity , Nicotinamide Phosphoribosyltransferase/metabolism , Oxidative Stress , Tuberculosis/immunology , Zebrafish/immunology , Animals , Apoptosis , Blotting, Western , Cell Adhesion , Cell Movement , Cell Proliferation , Cell Wall/metabolism , Cells, Cultured , Female , Host-Pathogen Interactions , Humans , Macrophages/cytology , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Microbial Viability , Mycobacterium marinum/growth & development , Mycobacterium tuberculosis/growth & development , NF-kappa B , Nicotinamide Phosphoribosyltransferase/genetics , Phagocytosis , Protein Conformation , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Tuberculosis/metabolism , Tuberculosis/microbiology , Virulence/immunology , Zebrafish/metabolism , Zebrafish/microbiology
8.
PLoS Pathog ; 10(6): e1004190, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24968056

ABSTRACT

Tuberculosis is still a major health problem worldwide. Currently it is not known what kind of immune responses lead to successful control and clearance of Mycobacterium tuberculosis. This gap in knowledge is reflected by the inability to develop sufficient diagnostic and therapeutic tools to fight tuberculosis. We have used the Mycobacterium marinum infection model in the adult zebrafish and taken advantage of heterogeneity of zebrafish population to dissect the characteristics of adaptive immune responses, some of which are associated with well-controlled latency or bacterial clearance while others with progressive infection. Differences in T cell responses between subpopulations were measured at the transcriptional level. It was discovered that a high total T cell level was usually associated with lower bacterial loads alongside with a T helper 2 (Th2)-type gene expression signature. At late time points, spontaneous reactivation with apparent symptoms was characterized by a low Th2/Th1 marker ratio and a substantial induction of foxp3 reflecting the level of regulatory T cells. Characteristic gata3/tbx21 has potential as a biomarker for the status of mycobacterial disease.


Subject(s)
Adaptive Immunity , Disease Models, Animal , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium marinum/immunology , Th2 Cells/immunology , Zebrafish/immunology , Algorithms , Animals , Animals, Genetically Modified , Bacterial Load , Biomarkers/blood , Biomarkers/metabolism , Disease Progression , Forkhead Transcription Factors/blood , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , GATA3 Transcription Factor/blood , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation , Lymphocyte Count , Lymphopoiesis , Microbial Viability , Mutation , Mycobacterium Infections, Nontuberculous/blood , Mycobacterium Infections, Nontuberculous/metabolism , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/growth & development , Mycobacterium marinum/isolation & purification , T-Box Domain Proteins/blood , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/microbiology , Th1 Cells/pathology , Th2 Cells/metabolism , Th2 Cells/microbiology , Th2 Cells/pathology , Up-Regulation , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish/microbiology , Zebrafish Proteins/blood , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Cell Microbiol ; 17(9): 1332-49, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25772333

ABSTRACT

Lipid droplets exist in virtually every cell type, ranging not only from mammals to plants, but also to eukaryotic and prokaryotic unicellular organisms such as Dictyostelium and bacteria. They serve among other roles as energy reservoir that cells consume in times of starvation. Mycobacteria and some other intracellular pathogens hijack these organelles as a nutrient source and to build up their own lipid inclusions. The mechanisms by which host lipid droplets are captured by the pathogenic bacteria are extremely poorly understood. Using the powerful Dictyostelium discoideum/Mycobacterium marinum infection model, we observed that, immediately after their uptake, lipid droplets translocate to the vicinity of the vacuole containing live but not dead mycobacteria. Induction of lipid droplets in Dictyostelium prior to infection resulted in a vast accumulation of neutral lipids and sterols inside the bacterium-containing compartment. Subsequently, under these conditions, mycobacteria accumulated much larger lipid inclusions. Strikingly, the Dictyostelium homologue of perilipin and the murine perilipin 2 surrounded bacteria that had escaped to the cytosol of Dictyostelium or microglial BV-2 cells respectively. Moreover, bacterial growth was inhibited in Dictyostelium plnA knockout cells. In summary, our results provide evidence that mycobacteria actively manipulate the lipid metabolism of the host from very early infection stages.


Subject(s)
Dictyostelium/metabolism , Dictyostelium/microbiology , Lipid Droplets/metabolism , Mycobacterium marinum/growth & development , Animals , Cell Line , Host-Pathogen Interactions , Mice , Microglia/metabolism , Microglia/microbiology , Models, Biological
10.
Wei Sheng Wu Xue Bao ; 56(9): 1496-1503, 2016 Sep.
Article in Zh | MEDLINE | ID: mdl-29738222

ABSTRACT

Objective: To identify differentially expressed proteins in Mycobacterium marinum wild-type (WT) and mkl::Tn mutant strains, and provide new clues for exploring the functions of mkl gene. Methods: Cellular proteins were extracted from cultures of M. marinum WT and mkl::Tn strains, and labelled with isobaric tags for relative and absolute quantitation (iTRAQ) 4-plex. Differentially expressed proteins were identified with LC-MS/MS and subjected to biological information analysis. Results: A total of 566 differentially expressed proteins were revealed, among which 232 proteins were up-regulated (ratio≥1.4) and 334 proteins were down-regulated (ratio≤0.7). These proteins are mainly associated with lipid metabolism, cell wall and cell processes, intermediary metabolism and respiration, and hypothetical proteins. The most down-regulated protein DesA3, is a fatty acid desaturase and involved in the synthesis of oleic acid. Further experiments showed that the growth of mkl::Tn strain was attenuated on 7H10-ADC agar plate without oleic acid, suggesting that mkl may play a role in the biosynthesis of oleic acid. Conclusion: Differentially expressed proteins were identified in M. marinum mkl::Tn compared to WT, and these results shed light on the mechanisms of mkl gene in mycobacterial pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Mycobacterium marinum/metabolism , Bacterial Proteins/genetics , Chromatography, Liquid , Mycobacterium marinum/chemistry , Mycobacterium marinum/genetics , Mycobacterium marinum/growth & development , Oleic Acid/biosynthesis , Proteomics , Tandem Mass Spectrometry
11.
Infect Immun ; 83(5): 1778-88, 2015 May.
Article in English | MEDLINE | ID: mdl-25690095

ABSTRACT

The interaction of environmental bacteria with unicellular eukaryotes is generally considered a major driving force for the evolution of intracellular pathogens, allowing them to survive and replicate in phagocytic cells of vertebrate hosts. To test this hypothesis on a genome-wide level, we determined for the intracellular pathogen Mycobacterium marinum whether it uses conserved strategies to exploit host cells from both protozoan and vertebrate origin. Using transposon-directed insertion site sequencing (TraDIS), we determined differences in genetic requirements for survival and replication in phagocytic cells of organisms from different kingdoms. In line with the general hypothesis, we identified a number of general virulence mechanisms, including the type VII protein secretion system ESX-1, biosynthesis of polyketide lipids, and utilization of sterols. However, we were also able to show that M. marinum contains an even larger set of host-specific virulence determinants, including proteins involved in the modification of surface glycolipids and, surprisingly, the auxiliary proteins of the ESX-1 system. Several of these factors were in fact counterproductive in other hosts. Therefore, M. marinum contains different sets of virulence factors that are tailored for specific hosts. Our data imply that although amoebae could function as a training ground for intracellular pathogens, they do not fully prepare pathogens for crossing species barriers.


Subject(s)
Genome, Bacterial , Microbial Viability , Mutagenesis, Insertional , Mycobacterium marinum/genetics , Mycobacterium marinum/physiology , Virulence Factors/metabolism , Acanthamoeba castellanii/microbiology , Animals , DNA Transposable Elements , Dictyostelium/microbiology , Humans , Mycobacterium marinum/growth & development , Phagocytes/microbiology , Virulence , Virulence Factors/genetics
12.
Infect Immun ; 83(7): 2844-54, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25939506

ABSTRACT

LytR-CpsA-Psr family proteins play an important role in bacterial cell wall integrity. Although the pathogenic relevance of LytR-CpsA-Psr family proteins has been studied in a few bacterial pathogens, their function in mycobacteria remains uncharacterized. In this work, a transposon insertion mutant (cpsA::Tn) of Mycobacterium marinum was studied. We found that inactivation of CpsA altered bacterial colony morphology, sliding motility, cell surface hydrophobicity, and cell wall permeability. Besides, the cpsA mutant exhibited a decreased arabinogalactan content, indicating that CpsA plays a role in cell wall assembly. Moreover, the mutant shows impaired growth within macrophage cell lines and is severely attenuated in zebrafish larvae and adult zebrafish. Taken together, our results indicated that CpsA, a previously uncharacterized protein, is important for mycobacterial cell wall integrity and is required for mycobacterial virulence.


Subject(s)
Bacterial Proteins/metabolism , Cell Wall/physiology , Mycobacterium marinum/physiology , Animals , Bacterial Proteins/genetics , Cell Line , Cell Wall/chemistry , Cell Wall/metabolism , DNA Transposable Elements , Hydrophobic and Hydrophilic Interactions , Larva/microbiology , Locomotion , Macrophages/immunology , Macrophages/microbiology , Mice , Mutagenesis, Insertional , Mycobacterium marinum/growth & development , Mycobacterium marinum/metabolism , Mycobacterium marinum/pathogenicity , Permeability , Virulence , Zebrafish/microbiology
13.
Immunogenetics ; 67(3): 135-47, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25503064

ABSTRACT

We present a RNA deep sequencing (RNAseq) analysis of a comparison of the transcriptome responses to infection of zebrafish larvae with Staphylococcus epidermidis and Mycobacterium marinum bacteria. We show how our developed GeneTiles software can improve RNAseq analysis approaches by more confidently identifying a large set of markers upon infection with these bacteria. For analysis of RNAseq data currently, software programs such as Bowtie2 and Samtools are indispensable. However, these programs that are designed for a LINUX environment require some dedicated programming skills and have no options for visualisation of the resulting mapped sequence reads. Especially with large data sets, this makes the analysis time consuming and difficult for non-expert users. We have applied the GeneTiles software to the analysis of previously published and newly obtained RNAseq datasets of our zebrafish infection model, and we have shown the applicability of this approach also to published RNAseq datasets of other organisms by comparing our data with a published mammalian infection study. In addition, we have implemented the DEXSeq module in the GeneTiles software to identify genes, such as glucagon A, that are differentially spliced under infection conditions. In the analysis of our RNAseq data, this has led to the possibility to improve the size of data sets that could be efficiently compared without using problem-dedicated programs, leading to a quick identification of marker sets. Therefore, this approach will also be highly useful for transcriptome analyses of other organisms for which well-characterised genomes are available.


Subject(s)
Fish Diseases/genetics , Fish Proteins/genetics , Mycobacterium Infections, Nontuberculous/veterinary , Software , Staphylococcal Infections/veterinary , Zebrafish/genetics , Alternative Splicing , Animals , Disease Models, Animal , Fish Diseases/microbiology , Gene Expression Profiling , Glucagon/genetics , High-Throughput Nucleotide Sequencing , Host-Pathogen Interactions , Larva/genetics , Larva/microbiology , Metabolic Networks and Pathways , Molecular Sequence Annotation , Mycobacterium Infections, Nontuberculous/genetics , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/growth & development , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/growth & development , Transcriptome , Zebrafish/microbiology
14.
PLoS Pathog ; 8(1): e1002487, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22253599

ABSTRACT

Multi-drug therapy is the standard-of-care treatment for tuberculosis. Despite this, virtually all studies of the pharmacodynamics (PD) of mycobacterial drugs employed for the design of treatment protocols are restricted to single agents. In this report, mathematical models and in vitro experiments with Mycobacterium marinum and five antimycobacterial drugs are used to quantitatively evaluate the pharmaco-, population and evolutionary dynamics of two-drug antimicrobial chemotherapy regimes. Time kill experiments with single and pairs of antibiotics are used to estimate the parameters and evaluate the fit of Hill-function-based PD models. While Hill functions provide excellent fits for the PD of each single antibiotic studied, rifampin, amikacin, clarithromycin, streptomycin and moxifloxacin, two-drug Hill functions with a unique interaction parameter cannot account for the PD of any of the 10 pairs of these drugs. If we assume two antibiotic-concentration dependent functions for the interaction parameter, one for sub-MIC and one for supra-MIC drug concentrations, the modified biphasic Hill function provides a reasonably good fit for the PD of all 10 pairs of antibiotics studied. Monte Carlo simulations of antibiotic treatment based on the experimentally-determined PD functions are used to evaluate the potential microbiological efficacy (rate of clearance) and evolutionary consequences (likelihood of generating multi-drug resistance) of these different drug combinations as well as their sensitivity to different forms of non-adherence to therapy. These two-drug treatment simulations predict varying outcomes for the different pairs of antibiotics with respect to the aforementioned measures of efficacy. In summary, Hill functions with biphasic drug-drug interaction terms provide accurate analogs for the PD of pairs of antibiotics and M. marinum. The models, experimental protocols and computer simulations used in this study can be applied to evaluate the potential microbiological and evolutionary efficacy of two-drug therapy for any bactericidal antibiotics and bacteria that can be cultured in vitro.


Subject(s)
Anti-Infective Agents/administration & dosage , Models, Theoretical , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium marinum/drug effects , Amikacin/administration & dosage , Amikacin/pharmacokinetics , Anti-Infective Agents/pharmacokinetics , Clarithromycin/administration & dosage , Clarithromycin/pharmacokinetics , Computer Simulation , Dose-Response Relationship, Drug , Drug Combinations , Drug Interactions/physiology , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/physiology , Humans , Microbial Sensitivity Tests , Models, Biological , Mycobacterium Infections, Nontuberculous/metabolism , Mycobacterium marinum/growth & development , Mycobacterium marinum/physiology , Rifampin/administration & dosage , Rifampin/pharmacokinetics , Tuberculosis/drug therapy , Tuberculosis/metabolism
15.
Adv Exp Med Biol ; 783: 251-66, 2013.
Article in English | MEDLINE | ID: mdl-23468113

ABSTRACT

Tuberculosis is characterized by the formation of complex immune cell aggregates called granulomas, which for nearly a century have been viewed as critical host-beneficial structures to restrict bacterial growth and spread. A different view has now emerged from real-time visualization of granuloma formation and its consequences in the optically transparent and genetically tractable zebrafish larva. Pathogenic mycobacteria have developed mechanisms to use host granulomas for their expansion and dissemination, at least during the innate phases of infection. Host processes that are intended to be beneficial-death of infected macrophages and their subsequent phagocytosis by macrophages that are newly recruited to the growing granuloma-are harnessed by mycobacteria for their own benefit. Mycobacteria can also render the granuloma a safe-haven in the more advanced stages of infection. An understanding of the host and bacterial pathways involved in tuberculous granuloma formation may suggest new ways to combat mycobacterial infection.


Subject(s)
Disease Models, Animal , Immune Evasion/immunology , Mycobacterium Infections, Nontuberculous/veterinary , Mycobacterium marinum/immunology , Tuberculoma/pathology , Zebrafish/immunology , Animals , Antigens, Bacterial/physiology , Apoptosis , BCG Vaccine/immunology , Bacterial Proteins/physiology , Disease Resistance , Enzyme Induction , Humans , Larva , Macrophages/microbiology , Matrix Metalloproteinase 9/physiology , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/pathology , Mycobacterium marinum/growth & development , Mycobacterium marinum/pathogenicity , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Necrosis , Tuberculoma/immunology , Tuberculoma/microbiology , Virulence , Zebrafish/growth & development , Zebrafish Proteins/physiology
16.
PLoS Pathog ; 6(5): e1000895, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20463815

ABSTRACT

The Esx-1 (type VII) secretion system is a major virulence determinant of pathogenic mycobacteria, including Mycobacterium marinum. However, the molecular events and host-pathogen interactions underlying Esx-1-mediated virulence in vivo remain unclear. Here we address this problem in a non-lethal mouse model of M. marinum infection that allows detailed quantitative analysis of disease progression. M. marinum established local infection in mouse tails, with Esx-1-dependent formation of caseating granulomas similar to those formed in human tuberculosis, and bone deterioration reminiscent of skeletal tuberculosis. Analysis of tails infected with wild type or Esx-1-deficient bacteria showed that Esx-1 enhanced generation of proinflammatory cytokines, including the secreted form of IL-1beta, suggesting that Esx-1 promotes inflammasome activation in vivo. In vitro experiments indicated that Esx-1-dependent inflammasome activation required the host NLRP3 and ASC proteins. Infection of wild type and ASC-deficient mice demonstrated that Esx-1-dependent inflammasome activation exacerbated disease without restricting bacterial growth, indicating a host-detrimental role of this inflammatory pathway in mycobacterial infection. These findings define an immunoregulatory role for Esx-1 in a specific host-pathogen interaction in vivo, and indicate that the Esx-1 secretion system promotes disease and inflammation through its ability to activate the inflammasome.


Subject(s)
Bacterial Proteins/immunology , Host-Pathogen Interactions/immunology , Macrophages/microbiology , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium marinum/growth & development , Animals , Apoptosis Regulatory Proteins , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Female , Inflammation/immunology , Inflammation/microbiology , Interleukin-1beta/metabolism , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/immunology , Mycobacterium marinum/pathogenicity , NLR Family, Pyrin Domain-Containing 3 Protein , Phagosomes/immunology , Tail/microbiology , Tuberculoma/immunology , Tuberculoma/microbiology , Virulence , Virulence Factors/genetics , Virulence Factors/immunology , Virulence Factors/metabolism
17.
Cell Microbiol ; 13(2): 246-58, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21040356

ABSTRACT

Bacterial ingestion and killing by phagocytic cells are essential processes to protect the human body from infectious microorganisms. However, only few proteins implicated in intracellular bacterial killing have been identified to date. We used Dictyostelium discoideum, a phagocytic bacterial predator, to study intracellular killing. In a random genetic screen we identified Kil2, a type V P-ATPase as an essential element for efficient intracellular killing of Klebsiella pneumoniae bacteria. Interestingly, kil2 knockout cells still killed efficiently several other species of bacteria, and did not show enhanced susceptibility to Mycobacterium marinum intracellular replication. Kil2 is present in the phagosomal membrane, and its structure suggests that it pumps cations into the phagosomal lumen. The killing defect of kil2 knockout cells was rescued by the addition of magnesium ions, suggesting that Kil2 may function as a magnesium pump. In agreement with this, kil2 mutant cells exhibited a specific defect for growth at high concentrations of magnesium. Phagosomal protease activity was lower in kil2 mutant cells than in wild-type cells, a phenotype reversed by the addition of magnesium to the medium. Kil2 may act as a magnesium pump maintaining magnesium concentration in phagosomes, thus ensuring optimal activity of phagosomal proteases and efficient killing of bacteria.


Subject(s)
Adenosine Triphosphatases/metabolism , Dictyostelium/microbiology , Klebsiella pneumoniae/growth & development , Magnesium/metabolism , Microbial Viability/drug effects , Mycobacterium marinum/growth & development , Phagosomes/microbiology , Dictyostelium/metabolism , Klebsiella pneumoniae/drug effects , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mycobacterium marinum/drug effects , Phagosomes/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
18.
PLoS One ; 17(1): e0262241, 2022.
Article in English | MEDLINE | ID: mdl-34986163

ABSTRACT

O-methylation of small molecules is a common modification widely present in most organisms. Type III polyketides undergo O-methylation at hydroxyl end to play a wide spectrum of roles in bacteria, plants, algae, and fungi. Mycobacterium marinum harbours a distinctive genomic cluster with a type III pks gene and genes for several polyketide modifiers including a methyltransferase gene, mmar_2193. This study reports functional analyses of MMAR_2193 and reveals multi-methylating potential of the protein. Comparative sequence analyses revealed conservation of catalytically important motifs in MMAR_2193 protein. Homology-based structure-function and molecular docking studies suggested type III polyketide cores as possible substrates for MMAR_2193 catalysis. In vitro enzymatic characterization revealed the capability of MMAR_2193 protein to utilize diverse polyphenolic substrates to methylate several hydroxyl positions on a single substrate molecule. High-resolution mass spectrometric analyses identified multi-methylations of type III polyketides in cell-free reconstitution assays. Notably, our metabolomics analyses identified some of these methylated molecules in biofilms of wild type Mycobacterium marinum. This study characterizes a novel mycobacterial O-methyltransferase protein with multi-methylating enzymatic ability that could be exploited to generate a palette of structurally distinct bioactive molecules.


Subject(s)
Methyltransferases/genetics , Methyltransferases/metabolism , Mycobacterium marinum/growth & development , Polyketides/chemistry , Amino Acid Motifs , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms , Cloning, Molecular , Conserved Sequence , Mass Spectrometry , Metabolomics , Methylation , Methyltransferases/chemistry , Models, Molecular , Molecular Docking Simulation , Mycobacterium marinum/enzymology , Mycobacterium marinum/genetics , Protein Conformation , Structural Homology, Protein
19.
J Clin Microbiol ; 49(7): 2691-3, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21543566

ABSTRACT

The reliability of the BacT/Alert 3D unit for automated detection of nontuberculous mycobacteria (NTM) that grow optimally at 30 °C was assessed. This system reliably maintained a temperature of 30 °C and detected 50% of the clinical NTM strains (5 Mycobacterium marinum and 3 Mycobacterium gordonae strains) faster than 37 °C culture.


Subject(s)
Automation/methods , Bacteriological Techniques/methods , Mycobacterium Infections/diagnosis , Mycobacterium marinum/classification , Mycobacterium marinum/isolation & purification , Nontuberculous Mycobacteria/classification , Nontuberculous Mycobacteria/isolation & purification , Humans , Mycobacterium marinum/growth & development , Nontuberculous Mycobacteria/growth & development , Sensitivity and Specificity , Temperature
20.
Methods Mol Biol ; 2314: 183-203, 2021.
Article in English | MEDLINE | ID: mdl-34235653

ABSTRACT

The Dictyostelium discoideum-Mycobacterium marinum host-pathogen system is a well-established and powerful alternative model system to study mycobacterial infections. In this chapter, we will describe three microscopy methods that allow the precise identification and quantification of very diverse phenotypes arising during infection of D. discoideum with M. marinum. First, at the lowest end of the scale, we use the InfectChip, a microfluidic device that enables the long-term monitoring of the integrated history of the infection course at the single-cell level. We use single-cell analysis to precisely map and quantitate the various fates of the host and the pathogen during infection. Second, a high-content microscopy setup was established to study the infection dynamics with high-throughput imaging of a large number of cells at the different critical stages of infection. The large datasets are then fed into a deep image analysis pipeline allowing the development of complex phenotypic analyses. Finally, as part of its life cycle, single D. discoideum amoebae aggregate by chemotaxis to form multicellular structures, which represent ordered assemblies of hundreds of thousands of cells. This transition represents a challenge for the monitoring of infection at multiple scales, from single cells to a true multicellular organism. In order to visualize and quantitate the fates of host cells and bacteria during the developmental cycle in a controlled manner, we can adjust the proportion of infected cells using live FAC-sorting. Then, cells are plated in defined humidity conditions on optical glass plates in order to image large fields, using tile scans, with the help of a spinning disc confocal microscope.


Subject(s)
Dictyostelium/microbiology , Host-Pathogen Interactions , Lab-On-A-Chip Devices , Microscopy, Electron/methods , Mycobacterium Infections, Nontuberculous/diagnosis , Mycobacterium marinum/growth & development , Single-Cell Analysis/methods , Dictyostelium/ultrastructure , Mycobacterium Infections, Nontuberculous/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL