Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
mSystems ; 6(3): e0050021, 2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34156290

ABSTRACT

The complex cell wall and biofilm matrix (ECM) act as key barriers to antibiotics in mycobacteria. Here, the ECM and envelope proteins of Mycobacterium marinum ATCC 927, a nontuberculous mycobacterial model, were monitored over 3 months by label-free proteomics and compared with cell surface proteins on planktonic cells to uncover pathways leading to virulence, tolerance, and persistence. We show that ATCC 927 forms pellicle-type and submerged-type biofilms (PBFs and SBFs, respectively) after 2 weeks and 2 days of growth, respectively, and that the increased CelA1 synthesis in this strain prevents biofilm formation and leads to reduced rifampicin tolerance. The proteomic data suggest that specific changes in mycolic acid synthesis (cord factor), Esx1 secretion, and cell wall adhesins explain the appearance of PBFs as ribbon-like cords and SBFs as lichen-like structures. A subpopulation of cells resisting 64× MIC rifampicin (persisters) was detected in both biofilm subtypes and already in 1-week-old SBFs. The key forces boosting their development could include subtype-dependent changes in asymmetric cell division, cell wall biogenesis, tricarboxylic acid/glyoxylate cycle activities, and energy/redox/iron metabolisms. The effect of various ambient oxygen tensions on each cell type and nonclassical protein secretion are likely factors explaining the majority of the subtype-specific changes. The proteomic findings also imply that Esx1-type protein secretion is more efficient in planktonic (PL) and PBF cells, while SBF may prefer both the Esx5 and nonclassical pathways to control virulence and prolonged viability/persistence. In conclusion, this study reports the first proteomic insight into aging mycobacterial biofilm ECMs and indicates biofilm subtype-dependent mechanisms conferring increased adaptive potential and virulence of nontuberculous mycobacteria. IMPORTANCE Mycobacteria are naturally resilient, and mycobacterial infections are notoriously difficult to treat with antibiotics, with biofilm formation being the main factor complicating the successful treatment of tuberculosis (TB). The present study shows that nontuberculous Mycobacterium marinum ATCC 927 forms submerged- and pellicle-type biofilms with lichen- and ribbon-like structures, respectively, as well as persister cells under the same conditions. We show that both biofilm subtypes differ in terms of virulence-, tolerance-, and persistence-conferring activities, highlighting the fact that both subtypes should be targeted to maximize the power of antimycobacterial treatment therapies.

2.
J Vis Exp ; (140)2018 10 08.
Article in English | MEDLINE | ID: mdl-30346391

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.


Subject(s)
Disease Models, Animal , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/physiology , Zebrafish , Animals , Gene Expression , Host-Pathogen Interactions , Mycobacterium Infections, Nontuberculous/genetics , Mycobacterium marinum/genetics , Real-Time Polymerase Chain Reaction
3.
Dis Model Mech ; 11(1)2018 01 29.
Article in English | MEDLINE | ID: mdl-29208761

ABSTRACT

Mycobacterium tuberculosis remains one of the most problematic infectious agents, owing to its highly developed mechanisms to evade host immune responses combined with the increasing emergence of antibiotic resistance. Host-directed therapies aiming to optimize immune responses to improve bacterial eradication or to limit excessive inflammation are a new strategy for the treatment of tuberculosis. In this study, we have established a zebrafish-Mycobacterium marinum natural host-pathogen model system to study induced protective immune responses in mycobacterial infection. We show that priming adult zebrafish with heat-killed Listeria monocytogenes (HKLm) at 1 day prior to M. marinum infection leads to significantly decreased mycobacterial loads in the infected zebrafish. Using rag1-/- fish, we show that the protective immunity conferred by HKLm priming can be induced through innate immunity alone. At 24 h post-infection, HKLm priming leads to a significant increase in the expression levels of macrophage-expressed gene 1 (mpeg1), tumor necrosis factor α (tnfa) and nitric oxide synthase 2b (nos2b), whereas superoxide dismutase 2 (sod2) expression is downregulated, implying that HKLm priming increases the number of macrophages and boosts intracellular killing mechanisms. The protective effects of HKLm are abolished when the injected material is pretreated with nucleases or proteinase K. Importantly, HKLm priming significantly increases the frequency of clearance of M. marinum infection by evoking sterilizing immunity (25 vs 3.7%, P=0.0021). In this study, immune priming is successfully used to induce sterilizing immunity against mycobacterial infection. This model provides a promising new platform for elucidating the mechanisms underlying sterilizing immunity and to develop host-directed treatment or prevention strategies against tuberculosis.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Cross-Priming/immunology , Immunity, Innate , Listeria monocytogenes/physiology , Mycobacterium tuberculosis/immunology , Sterilization , Tuberculosis/immunology , Tuberculosis/microbiology , Zebrafish/microbiology , Aging , Animals , Bacterial Load , Bacterial Proteins/metabolism , Disease Models, Animal , Down-Regulation , Female , Hot Temperature , Larva , Macrophages/microbiology , Male , Mycobacterium marinum/immunology , Nucleic Acids/metabolism , Oxygen Consumption , Tuberculosis/prevention & control , Zebrafish Proteins/metabolism
4.
PLoS Pathog ; 12(7): e1005746, 2016 07.
Article in English | MEDLINE | ID: mdl-27414410

ABSTRACT

Cellular immune responses require the generation and recruitment of diverse blood cell types that recognize and kill pathogens. In Drosophila melanogaster larvae, immune-inducible lamellocytes participate in recognizing and killing parasitoid wasp eggs. However, the sequence of events required for lamellocyte generation remains controversial. To study the cellular immune system, we developed a flow cytometry approach using in vivo reporters for lamellocytes as well as for plasmatocytes, the main hemocyte type in healthy larvae. We found that two different blood cell lineages, the plasmatocyte and lamellocyte lineages, contribute to the generation of lamellocytes in a demand-adapted hematopoietic process. Plasmatocytes transdifferentiate into lamellocyte-like cells in situ directly on the wasp egg. In parallel, a novel population of infection-induced cells, which we named lamelloblasts, appears in the circulation. Lamelloblasts proliferate vigorously and develop into the major class of circulating lamellocytes. Our data indicate that lamellocyte differentiation upon wasp parasitism is a plastic and dynamic process. Flow cytometry with in vivo hemocyte reporters can be used to study this phenomenon in detail.


Subject(s)
Cell Proliferation , Cell Transdifferentiation/physiology , Drosophila melanogaster/physiology , Drosophila melanogaster/parasitology , Hematopoiesis/physiology , Hemocytes/cytology , Wasps , Animals , Cell Lineage , Flow Cytometry/methods , Immunohistochemistry , Larva , Microscopy, Confocal
5.
Immunol Lett ; 170: 42-51, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26730849

ABSTRACT

Cytokines are a large and diverse group of small proteins that can affect many biological processes, but most commonly cytokines are known as mediators of the immune response. In the event of an infection, cytokines are produced in response to an immune stimulus, and they function as key regulators of the immune response. Cytokines come in many shapes and sizes, and although they vary greatly in structure, their functions have been well conserved in evolution. The immune signaling pathways that respond to cytokines are remarkably conserved from fly to man. Therefore, Drosophila melanogaster, provides an excellent platform for studying the biology and function of cytokines. In this review, we will describe the cytokines and cytokine-like molecules found in the fly and discuss their roles in host immunity.


Subject(s)
Cytokines/metabolism , Drosophila/immunology , Drosophila/metabolism , Immunity , Animals , Cytokines/genetics , Drosophila/genetics , Immune System/cytology , Immune System/physiology , Signal Transduction
6.
PLoS Pathog ; 11(5): e1004895, 2015 May.
Article in English | MEDLINE | ID: mdl-25965263

ABSTRACT

The cellular immune response against parasitoid wasps in Drosophila involves the activation, mobilization, proliferation and differentiation of different blood cell types. Here, we have assessed the role of Edin (elevated during infection) in the immune response against the parasitoid wasp Leptopilina boulardi in Drosophila melanogaster larvae. The expression of edin was induced within hours after a wasp infection in larval fat bodies. Using tissue-specific RNAi, we show that Edin is an important determinant of the encapsulation response. Although edin expression in the fat body was required for the larvae to mount a normal encapsulation response, it was dispensable in hemocytes. Edin expression in the fat body was not required for lamellocyte differentiation, but it was needed for the increase in plasmatocyte numbers and for the release of sessile hemocytes into the hemolymph. We conclude that edin expression in the fat body affects the outcome of a wasp infection by regulating the increase of plasmatocyte numbers and the mobilization of sessile hemocytes in Drosophila larvae.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Fat Body/metabolism , Host-Parasite Interactions , Up-Regulation , Wasps/immunology , Animals , Animals, Genetically Modified , Biomarkers/metabolism , Crosses, Genetic , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/immunology , Drosophila melanogaster/parasitology , Fat Body/cytology , Fat Body/immunology , Gene Knockdown Techniques , Genes, Reporter , Hematopoiesis, Extramedullary , Hemocytes/cytology , Hemocytes/immunology , Hemocytes/metabolism , Hemolymph/cytology , Hemolymph/immunology , Hemolymph/metabolism , Immunity, Innate , Kinetics , Larva/cytology , Larva/immunology , Larva/metabolism , Larva/parasitology , Ovum/immunology , Ovum/physiology , Parasite Egg Count , RNA Interference , Recombinant Fusion Proteins/metabolism , Wasps/physiology
7.
PLoS One ; 9(8): e102568, 2014.
Article in English | MEDLINE | ID: mdl-25102059

ABSTRACT

The Toll signaling pathway, first discovered in Drosophila, has a well-established role in immune responses in insects as well as in mammals. In Drosophila, the Toll-dependent induction of antimicrobial peptide production has been intensely studied as a model for innate immune responses in general. Besides this humoral immune response, Toll signaling is also known to activate blood cells in a reaction that is similar to the cellular immune response to parasite infections, but the mechanisms of this response are poorly understood. Here we have studied this response in detail, and found that Toll signaling in several different tissues can activate a cellular immune defense, and that this response does not require Toll signaling in the blood cells themselves. Like in the humoral immune response, we show that Toll signaling in the fat body (analogous to the liver in vertebrates) is of major importance in the Toll-dependent activation of blood cells. However, this Toll-dependent mechanism of blood cell activation contributes very little to the immune response against the parasitoid wasp, Leptopilina boulardi, probably because the wasp is able to suppress Toll induction. Other redundant pathways may be more important in the defense against this pathogen.


Subject(s)
Drosophila Proteins/physiology , Drosophila/immunology , Fat Body/metabolism , Hemocytes/metabolism , Host-Parasite Interactions/immunology , Immunity, Cellular , Larva/immunology , Toll-Like Receptors/physiology , Animals , Drosophila/metabolism , Drosophila/parasitology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Larva/metabolism , Larva/parasitology , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Wasps/physiology
8.
PLoS One ; 7(5): e37153, 2012.
Article in English | MEDLINE | ID: mdl-22606343

ABSTRACT

Drosophila is a well-established model organism for studying innate immunity because of its high resistance against microbial infections and lack of adaptive immunity. In addition, the immune signaling cascades found in Drosophila are evolutionarily conserved. Upon infection, activation of the immune signaling pathways, Toll and Imd, leads to the expression of multiple immune response genes, such as the antimicrobial peptides (AMPs). Previously, we identified an uncharacterized gene edin among the genes, which were strongly induced upon stimulation with Escherichia coli in Drosophila S2 cells. Edin has been associated with resistance against Listeria monocytogenes, but its role in Drosophila immunity remains elusive. In this study, we examined the role of Edin in the immune response of Drosophila both in vitro and in vivo. We report that edin expression is dependent on the Imd-pathway NF-κB transcription factor Relish and that it is expressed upon infection both in vitro and in vivo. Edin encodes a pro-protein, which is further processed in S2 cells. In our experiments, Edin did not bind microbes, nor did it possess antimicrobial activity to tested microbial strains in vitro or in vivo. Furthermore, edin RNAi did not significantly affect the expression of AMPs in vitro or in vivo. However, edin RNAi flies showed modestly impaired resistance to E. faecalis infection. We conclude that Edin has no potent antimicrobial properties but it appears to be important for E. faecalis infection via an uncharacterized mechanism. Further studies are still required to elucidate the exact role of Edin in the Drosophila immune response.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/immunology , Drosophila melanogaster/genetics , Drosophila melanogaster/immunology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Base Sequence , Cell Line , DNA Primers/genetics , Drosophila Proteins/antagonists & inhibitors , Drosophila melanogaster/microbiology , Genes, Insect , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/immunology , Immunity, Innate , Molecular Sequence Data , RNA Interference , Sequence Homology, Amino Acid , Signal Transduction , Species Specificity , Transcription Factors/genetics , Transcription Factors/immunology
9.
APMIS ; 119(10): 651-62, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21917002

ABSTRACT

In mammals, phagocytosis coordinates host defence on two levels: It acts both as an effector of the innate immunity, as well as an initiator of the adaptive immunity. The fruit fly Drosophila melanogaster (D. melanogaster) lacks the adaptive immune response, and the role of Drosophila plasmatocytes, cells that resemble phagocytosing mammalian macrophages, is limited to innate immune responses. During the past years, several studies have shed light on the role of phagocytosis in the Drosophila host defence. At least in some infection models, the systemic production of potent antimicrobial peptides (AMPs) does not completely compensate for the need for cellular immune responses. As a model, Drosophila offers powerful tools for studying phagocytosis including, large-scale RNA interference (RNAi) based in vitro screens that can be combined with classical Drosophila genetics. These kinds of approaches have led to important discoveries related especially to microbial recognition by Drosophila plasmatocytes. Events following initial recognition, however, have remained more elusive. This review summarizes the current knowledge on Drosophila phagocytosis focusing on the most recent advancements in the field, and highlighting the benefits the Drosophila system has to offer for research on phagocytosis.


Subject(s)
Drosophila melanogaster/immunology , Immunity, Innate/immunology , Phagocytosis/immunology , Animals , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Hemocytes/immunology , Host-Pathogen Interactions/immunology , Phagocytosis/genetics , Signal Transduction/immunology
10.
J Leukoc Biol ; 89(5): 649-59, 2011 May.
Article in English | MEDLINE | ID: mdl-21208897

ABSTRACT

Phagocytosis is an ancient cellular process that plays an important role in host defense. In Drosophila melanogaster phagocytic, macrophage-like hemocytes recognize and ingest microbes. We performed an RNAi-based in vitro screen in the Drosophila hemocyte cell line S2 and identified Abi, cpa, cofilin regulator 14-3-3ζ, tlk, CG2765, and CG15609 as mediators of bacterial phagocytosis. Of these identified genes, 14-3-3ζ had an evolutionarily conserved role in phagocytosis: bacterial phagocytosis was compromised when 14-3-3ζ was targeted with RNAi in primary Drosophila hemocytes and when the orthologous genes Ywhab and Ywhaz were silenced in zebrafish and mouse RAW 264.7 cells, respectively. In Drosophila and zebrafish infection models, 14-3-3ζ was required for resistance against Staphylococcus aureus. We conclude that 14-3-3ζ is essential for phagocytosis and microbial resistance in insects and vertebrates.


Subject(s)
14-3-3 Proteins/genetics , Actin Depolymerizing Factors/genetics , Biological Evolution , Drosophila melanogaster/genetics , Drosophila melanogaster/microbiology , Escherichia coli/pathogenicity , Phagocytosis/physiology , 14-3-3 Proteins/antagonists & inhibitors , Animals , Cells, Cultured , Drosophila melanogaster/immunology , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Female , Gene Silencing , Genes, Insect , Hemocytes/metabolism , Male , Mice , RNA, Small Interfering/genetics , Zebrafish/genetics , Zebrafish/immunology , Zebrafish/microbiology
11.
FASEB J ; 24(11): 4467-79, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20624926

ABSTRACT

JAK/STAT signaling pathway is evolutionarily conserved and tightly regulated. We carried out a reporter-based genome-wide RNAi in vitro screen to identify genes that regulate Drosophila JAK/STAT pathway and found 5 novel regulators. Of these, CG14225 is a negative regulator structurally related to the Drosophila JAK/STAT pathway receptor Domeless, especially in the extracellular domain, and to the mammalian IL-6 receptor and the signal transducer gp130. CG14225 coimmunoprecipitates with Domeless and its associated kinase hopscotch in S2 cells. CG14225 RNAi caused hyperphosphorylation of the transcription factor Stat92E in S2 cells on stimulation with the Drosophila JAK/STAT pathway ligand unpaired. CG14225 RNAi in vivo hyperactivated JAK/STAT target genes on septic injury and enhanced unpaired-induced eye overgrowth, and was thus named the eye transformer (ET). In the gastrointestinal infection model, where JAK/STAT signaling is important for stem cell renewal, CG14225/ET RNAi was protective in vivo. In conclusion, we have identified ET as a novel negative regulator of the Drosophila JAK/STAT pathway both in vitro and in vivo, and it functions in regulating Stat92E phosphorylation.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Animals , Cell Line , Cells, Cultured , Disease Models, Animal , Drosophila melanogaster/growth & development , Drosophila melanogaster/microbiology , Eye/growth & development , Gene Expression Regulation , Genome-Wide Association Study , Luciferases/metabolism , Phosphorylation , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Serratia marcescens/physiology
12.
J Immunol ; 180(8): 5413-22, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18390723

ABSTRACT

NF-kappaB transcription factors are involved in evolutionarily conserved signaling pathways controlling multiple cellular processes including apoptosis and immune and inflammatory responses. Immune response of the fruit fly Drosophila melanogaster to Gram-negative bacteria is primarily mediated via the Imd (immune deficiency) pathway, which closely resembles the mammalian TNFR signaling pathway. Instead of cytokines, the main outcome of Imd signaling is the production of antimicrobial peptides. The pathway activity is delicately regulated. Although many of the Imd pathway components are known, the mechanisms of negative regulation are more elusive. In this study we report that a previously uncharacterized gene, pirk, is highly induced upon Gram-negative bacterial infection in Drosophila in vitro and in vivo. pirk encodes a cytoplasmic protein that coimmunoprecipitates with Imd and the cytoplasmic tail of peptidoglycan recognition protein LC (PGRP-LC). RNA interference-mediated down-regulation of Pirk caused Imd pathway hyperactivation upon infection with Gram-negative bacteria, while overexpression of pirk reduced the Imd pathway response both in vitro and in vivo. Furthermore, pirk-overexpressing flies were more susceptible to Gram-negative bacterial infection than wild-type flies. We conclude that Pirk is a negative regulator of the Imd pathway.


Subject(s)
Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Drosophila melanogaster/microbiology , Enterobacter cloacae/immunology , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/immunology , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/immunology , Drosophila melanogaster/metabolism , Enterobacter cloacae/physiology , Genes, Insect , Molecular Sequence Data , NF-kappa B/immunology , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , RNA Interference , Sequence Alignment , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...