Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Int J Stroke ; : 17474930241242266, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38494462

ABSTRACT

BACKGROUND: Due to the rarity of cerebral venous thrombosis (CVT), performing high-quality scientific research in this field is challenging. Providing answers to unresolved research questions will improve prevention, diagnosis, and treatment, and ultimately translate to a better outcome of patients with CVT. We present an international research agenda, in which the most important research questions in the field of CVT are prioritized. AIMS: This research agenda has three distinct goals: (1) to provide inspiration and focus to research on CVT for the coming years, (2) to reinforce international collaboration, and (3) to facilitate the acquisition of research funding. SUMMARY OF REVIEW: This international research agenda is the result of a research summit organized by the International Cerebral Venous Thrombosis Consortium in Amsterdam, the Netherlands, in June 2023. The summit brought together 45 participants from 15 countries including clinical researchers from various disciplines, patients who previously suffered from CVT, and delegates from industry and non-profit funding organizations. The research agenda is categorized into six pre-specified themes: (1) epidemiology and clinical features, (2) life after CVT, (3) neuroimaging and diagnosis, (4) pathophysiology, (5) medical treatment, and (6) endovascular treatment. For each theme, we present two to four research questions, followed by a brief substantiation per question. The research questions were prioritized by the participants of the summit through consensus discussion. CONCLUSIONS: This international research agenda provides an overview of the most burning research questions on CVT. Answering these questions will advance our understanding and management of CVT, which will ultimately lead to improved outcomes for CVT patients worldwide.

2.
Biology (Basel) ; 12(6)2023 Jun 04.
Article in English | MEDLINE | ID: mdl-37372102

ABSTRACT

Existing drug treatment against tuberculosis is no match against the increasing number of multi-drug resistant strains of its causative agent, Mycobacterium tuberculosis (Mtb). A better understanding of how mycobacteria subvert the host immune defenses is crucial for developing novel therapeutic strategies. A potential approach is enhancing the activity of the autophagy machinery, which can direct bacteria to autophagolysosomal degradation. However, the interplay specifics between mycobacteria and the autophagy machinery must be better understood. Here, we analyzed live imaging data from the zebrafish model of tuberculosis to characterize mycobacteria-autophagy interactions during the early stages of infection in vivo. For high-resolution imaging, we microinjected fluorescent Mycobacterium marinum (Mm) into the tail fin tissue of zebrafish larvae carrying the GFP-LC3 autophagy reporter. We detected phagocytosed Mm clusters and LC3-positive Mm-containing vesicles within the first hour of infection. LC3 associations with these vesicles were transient and heterogeneous, ranging from simple vesicles to complex compound structures, dynamically changing shape by fusions between Mm-containing and empty vesicles. LC3-Mm-vesicles could adopt elongated shapes during cell migration or alternate between spacious and compact morphologies. LC3-Mm-vesicles were also observed in cells reverse migrating from the infection site, indicating that the autophagy machinery fails to control infection before tissue dissemination.

3.
Cells ; 12(6)2023 03 07.
Article in English | MEDLINE | ID: mdl-36980169

ABSTRACT

Damage-Regulated Autophagy Modulator 1 (DRAM1) is an infection-inducible membrane protein, whose function in the immune response is incompletely understood. Based on previous results in a zebrafish infection model, we have proposed that DRAM1 is a host resistance factor against intracellular mycobacterial infection. To gain insight into the cellular processes underlying DRAM1-mediated host defence, here we studied the interaction of DRAM1 with Mycobacterium marinum in murine RAW264.7 macrophages. We found that, shortly after phagocytosis, DRAM1 localised in a punctate pattern to mycobacteria, which gradually progressed to full DRAM1 envelopment of the bacteria. Within the same time frame, DRAM1-positive mycobacteria colocalised with the LC3 marker for autophagosomes and LysoTracker and LAMP1 markers for (endo)lysosomes. Knockdown analysis revealed that DRAM1 is required for the recruitment of LC3 and for the acidification of mycobacteria-containing vesicles. A reduction in the presence of LAMP1 further suggested reduced fusion of lysosomes with mycobacteria-containing vesicles. Finally, we show that DRAM1 knockdown impairs the ability of macrophages to defend against mycobacterial infection. Together, these results support that DRAM1 promotes the trafficking of mycobacteria through the degradative (auto)phagolysosomal pathway. Considering its prominent effect on host resistance to intracellular infection, DRAM1 is a promising target for therapeutic modulation of the microbicidal capacity of macrophages.


Subject(s)
Membrane Proteins , Mycobacterium Infections , Mycobacterium marinum , Animals , Mice , Autophagy , Lysosomes/metabolism , Macrophages/metabolism , Membrane Proteins/metabolism
4.
mBio ; 14(1): e0302422, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36475748

ABSTRACT

The global burden of tuberculosis (TB) is aggravated by the continuously increasing emergence of drug resistance, highlighting the need for innovative therapeutic options. The concept of host-directed therapy (HDT) as adjunctive to classical antibacterial therapy with antibiotics represents a novel and promising approach for treating TB. Here, we have focused on repurposing the clinically used anticancer drug tamoxifen, which was identified as a molecule with strong host-directed activity against intracellular Mycobacterium tuberculosis (Mtb). Using a primary human macrophage Mtb infection model, we demonstrate the potential of tamoxifen against drug-sensitive as well as drug-resistant Mtb bacteria. The therapeutic effect of tamoxifen was confirmed in an in vivo TB model based on Mycobacterium marinum infection of zebrafish larvae. Tamoxifen had no direct antimicrobial effects at the concentrations used, confirming that tamoxifen acted as an HDT drug. Furthermore, we demonstrate that the antimycobacterial effect of tamoxifen is independent of its well-known target the estrogen receptor (ER) pathway, but instead acts by modulating autophagy, in particular the lysosomal pathway. Through RNA sequencing and microscopic colocalization studies, we show that tamoxifen stimulates lysosomal activation and increases the localization of mycobacteria in lysosomes both in vitro and in vivo, while inhibition of lysosomal activity during tamoxifen treatment partly restores mycobacterial survival. Thus, our work highlights the HDT potential of tamoxifen and proposes it as a repurposed molecule for the treatment of TB. IMPORTANCE Tuberculosis (TB) is the world's most lethal infectious disease caused by a bacterial pathogen, Mycobacterium tuberculosis. This pathogen evades the immune defenses of its host and grows intracellularly in immune cells, particularly inside macrophages. There is an urgent need for novel therapeutic strategies because treatment of TB patients is increasingly complicated by rising antibiotic resistance. In this study, we explored a breast cancer drug, tamoxifen, as a potential anti-TB drug. We show that tamoxifen acts as a so-called host-directed therapeutic, which means that it does not act directly on the bacteria but helps the host macrophages combat the infection more effectively. We confirmed the antimycobacterial effect of tamoxifen in a zebrafish model for TB and showed that it functions by promoting the delivery of mycobacteria to digestive organelles, the lysosomes. These results support the high potential of tamoxifen to be repurposed to fight antibiotic-resistant TB infections by host-directed therapy.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Animals , Humans , Zebrafish , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Drug Repositioning , Tuberculosis/microbiology , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Mycobacterium tuberculosis/genetics
5.
Nucleic Acids Res ; 50(2): e10, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34734265

ABSTRACT

The interplay between three-dimensional chromosome organisation and genomic processes such as replication and transcription necessitates in vivo studies of chromosome dynamics. Fluorescent organic dyes are often used for chromosome labelling in vivo. The mode of binding of these dyes to DNA cause its distortion, elongation, and partial unwinding. The structural changes induce DNA damage and interfere with the binding dynamics of chromatin-associated proteins, consequently perturbing gene expression, genome replication, and cell cycle progression. We have developed a minimally-perturbing, genetically encoded fluorescent DNA label consisting of a (photo-switchable) fluorescent protein fused to the DNA-binding domain of H-NS - a bacterial nucleoid-associated protein. We show that this DNA label, abbreviated as HI-NESS (H-NS-based indicator for nucleic acid stainings), is minimally-perturbing to genomic processes and labels chromosomes in eukaryotic cells in culture, and in zebrafish embryos with preferential binding to AT-rich chromatin.


Subject(s)
Bacterial Proteins/metabolism , Biological Assay/methods , DNA, Bacterial/metabolism , DNA-Binding Proteins/metabolism , Staining and Labeling/methods , Animals , Bacterial Proteins/genetics , Cell Line , Cloning, Molecular , DNA Replication , DNA, Bacterial/chemistry , DNA-Binding Proteins/genetics , Fluorescent Dyes , Gene Expression , Genetic Vectors , Microscopy, Fluorescence
6.
Cells ; 9(11)2020 10 29.
Article in English | MEDLINE | ID: mdl-33138004

ABSTRACT

Modeling human infectious diseases using the early life stages of zebrafish provides unprecedented opportunities for visualizing and studying the interaction between pathogens and phagocytic cells of the innate immune system. Intracellular pathogens use phagocytes or other host cells, like gut epithelial cells, as a replication niche. The intracellular growth of these pathogens can be counteracted by host defense mechanisms that rely on the autophagy machinery. In recent years, zebrafish embryo infection models have provided in vivo evidence for the significance of the autophagic defenses and these models are now being used to explore autophagy as a therapeutic target. In line with studies in mammalian models, research in zebrafish has shown that selective autophagy mediated by ubiquitin receptors, such as p62, is important for host resistance against several bacterial pathogens, including Shigella flexneri, Mycobacterium marinum, and Staphylococcus aureus. Furthermore, an autophagy related process, Lc3-associated phagocytosis (LAP), proved host beneficial in the case of Salmonella Typhimurium infection but host detrimental in the case of S. aureus infection, where LAP delivers the pathogen to a replication niche. These studies provide valuable information for developing novel therapeutic strategies aimed at directing the autophagy machinery towards bacterial degradation.


Subject(s)
Autophagy , Bacterial Infections/metabolism , Bacterial Infections/pathology , Microtubule-Associated Proteins/metabolism , Phagocytosis , Zebrafish Proteins/metabolism , Animals , Bacteria/metabolism , Bacterial Infections/microbiology , Disease Models, Animal , Humans
7.
Microsc Microanal ; 26(5): 978-988, 2020 10.
Article in English | MEDLINE | ID: mdl-32878652

ABSTRACT

Symmetry is omnipresent in nature and we encounter symmetry routinely in our everyday life. It is also common on the microscopic level, where symmetry is often key to the proper function of core biological processes. The human brain is exquisitely well suited to recognize such symmetrical features with ease. In contrast, computational recognition of such patterns in images is still surprisingly challenging. In this paper we describe a mathematical approach to identifying smaller local symmetrical structures within larger images. Our algorithm attributes a local symmetry score to each image pixel, which subsequently allows the identification of the symmetrical centers of an object. Though there are already many methods available to detect symmetry in images, to the best of our knowledge, our algorithm is the first that is easily applicable in ImageJ/FIJI. We have created an interactive plugin in FIJI that allows the detection and thresholding of local symmetry values. The plugin combines the different reflection symmetry axis of a square to get a good coverage of reflection symmetry in all directions. To demonstrate the plugins potential, we analyzed images of bacterial chemoreceptor arrays and intracellular vesicle trafficking events, which are two prominent examples of biological systems with symmetrical patterns.


Subject(s)
Image Processing, Computer-Assisted/methods , Pattern Recognition, Automated , Physical Phenomena , Algorithms , Chemotaxis , Forensic Anthropology , Humans , Machine Learning
8.
Cell Death Dis ; 11(4): 277, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332700

ABSTRACT

DNA damage regulated autophagy modulator 1 (DRAM1) is a stress-inducible regulator of autophagy and cell death. DRAM1 has been implicated in cancer, myocardial infarction, and infectious diseases, but the molecular and cellular functions of this transmembrane protein remain poorly understood. Previously, we have proposed DRAM1 as a host resistance factor for tuberculosis (TB) and a potential target for host-directed anti-infective therapies. In this study, we generated a zebrafish dram1 mutant and investigated its loss-of-function effects during Mycobacterium marinum (Mm) infection, a widely used model in TB research. In agreement with previous knockdown analysis, dram1 mutation increased the susceptibility of zebrafish larvae to Mm infection. RNA sequencing revealed major effects of Dram1 deficiency on metabolic, immune response, and cell death pathways during Mm infection, and only minor effects on proteinase and metabolic pathways were found under uninfected conditions. Furthermore, unchallenged dram1 mutants did not display overt autophagic defects, but autophagic targeting of Mm was reduced in the absence of Dram1. The phagocytic ability of macrophages in dram1 mutants was unaffected, but acidification of Mm-containing vesicles was strongly reduced, indicating that Dram1 is required for phagosome maturation. By in vivo imaging, we observed that Dram1-deficient macrophages fail to restrict Mm during early stages of infection. The resulting increase in bacterial burden could be reverted by knockdown of inflammatory caspase a (caspa) and gasdermin Eb (gsdmeb), demonstrating pyroptosis as the mechanism underlying premature cell death of Mm-infected macrophages in dram1 mutants. Collectively, these data demonstrate that dissemination of mycobacterial infection in zebrafish larvae is promoted in the absence of Dram1 due to reduced maturation of mycobacteria-containing vesicles, failed intracellular containment, and consequent pyroptotic death of infected macrophages. These results provide new evidence that Dram1 plays a central role in host resistance to intracellular infection, acting at the crossroad of autophagy and cell death.


Subject(s)
Autophagy/genetics , Macrophages/metabolism , Membrane Proteins/deficiency , Mycobacterium Infections, Nontuberculous/metabolism , Pyroptosis/genetics , Tuberculosis/genetics , Animals , Cell Death , Humans , Zebrafish
9.
Cell Host Microbe ; 27(2): 238-248.e7, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31901518

ABSTRACT

Antimicrobial resistance in tuberculosis (TB) is a public health threat of global dimension, worsened by increasing drug resistance. Host-directed therapy (HDT) is an emerging concept currently explored as an adjunct therapeutic strategy for TB. One potential host target is the ligand-activated transcription factor aryl hydrocarbon receptor (AhR), which binds TB virulence factors and controls antibacterial responses. Here, we demonstrate that in the context of therapy, the AhR binds several TB drugs, including front line drugs rifampicin (RIF) and rifabutin (RFB), resulting in altered host defense and drug metabolism. AhR sensing of TB drugs modulates host defense mechanisms, notably impairs phagocytosis, and increases TB drug metabolism. Targeting AhR in vivo with a small-molecule inhibitor increases RFB-treatment efficacy. Thus, the AhR markedly impacts TB outcome by affecting both host defense and drug metabolism. As a corollary, we propose the AhR as a potential target for HDT in TB in adjunct to canonical chemotherapy.


Subject(s)
Antitubercular Agents/metabolism , Mycobacterium tuberculosis , Receptors, Aryl Hydrocarbon/drug effects , Tuberculosis/drug therapy , Animals , Antitubercular Agents/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Humans , Immunity, Cellular/drug effects , Mycobacterium marinum/drug effects , Mycobacterium marinum/pathogenicity , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/pathogenicity , Phagocytosis/drug effects , Receptors, Aryl Hydrocarbon/metabolism , Rifabutin/metabolism , Rifabutin/therapeutic use , Rifampin/metabolism , Rifampin/therapeutic use , THP-1 Cells , Treatment Outcome , Tuberculosis/microbiology , Zebrafish
10.
PLoS Pathog ; 15(2): e1007329, 2019 02.
Article in English | MEDLINE | ID: mdl-30818338

ABSTRACT

Mycobacterial pathogens are the causative agents of chronic infectious diseases like tuberculosis and leprosy. Autophagy has recently emerged as an innate mechanism for defense against these intracellular pathogens. In vitro studies have shown that mycobacteria escaping from phagosomes into the cytosol are ubiquitinated and targeted by selective autophagy receptors. However, there is currently no in vivo evidence for the role of selective autophagy receptors in defense against mycobacteria, and the importance of autophagy in control of mycobacterial diseases remains controversial. Here we have used Mycobacterium marinum (Mm), which causes a tuberculosis-like disease in zebrafish, to investigate the function of two selective autophagy receptors, Optineurin (Optn) and SQSTM1 (p62), in host defense against a mycobacterial pathogen. To visualize the autophagy response to Mm in vivo, optn and p62 zebrafish mutant lines were generated in the background of a GFP-Lc3 autophagy reporter line. We found that loss-of-function mutation of optn or p62 reduces autophagic targeting of Mm, and increases susceptibility of the zebrafish host to Mm infection. Transient knockdown studies confirmed the requirement of both selective autophagy receptors for host resistance against Mm infection. For gain-of-function analysis, we overexpressed optn or p62 by mRNA injection and found this to increase the levels of GFP-Lc3 puncta in association with Mm and to reduce the Mm infection burden. Taken together, our results demonstrate that both Optn and p62 are required for autophagic host defense against mycobacterial infection and support that protection against tuberculosis disease may be achieved by therapeutic strategies that enhance selective autophagy.


Subject(s)
Host-Pathogen Interactions/physiology , Mycobacterium Infections, Nontuberculous/metabolism , Mycobacterium marinum/pathogenicity , Animals , Animals, Genetically Modified , Autophagy/physiology , Cell Cycle Proteins , Disease Models, Animal , Humans , Macrophages , Membrane Transport Proteins , Mycobacterium/pathogenicity , Mycobacterium Infections/metabolism , Phagosomes , Sequestosome-1 Protein , Transcription Factor TFIIIA/metabolism , Tuberculosis , Ubiquitin , Zebrafish/metabolism , Zebrafish Proteins/metabolism
11.
Autophagy ; 15(5): 796-812, 2019 05.
Article in English | MEDLINE | ID: mdl-30676840

ABSTRACT

Innate immune defense against intracellular pathogens, like Salmonella, relies heavily on the autophagy machinery of the host. This response is studied intensively in epithelial cells, the target of Salmonella during gastrointestinal infections. However, little is known of the role that autophagy plays in macrophages, the predominant carriers of this pathogen during systemic disease. Here we utilize a zebrafish embryo model to study the interaction of S. enterica serovar Typhimurium with the macroautophagy/autophagy machinery of macrophages in vivo. We show that phagocytosis of live but not heat-killed Salmonella triggers recruitment of the autophagy marker GFP-Lc3 in a variety of patterns labeling tight or spacious bacteria-containing compartments, also revealed by electron microscopy. Neutrophils display similar GFP-Lc3 associations, but genetic modulation of the neutrophil/macrophage balance and ablation experiments show that macrophages are critical for the defense response. Deficiency of atg5 reduces GFP-Lc3 recruitment and impairs host resistance, in contrast to atg13 deficiency, indicating that Lc3-Salmonella association at this stage is independent of the autophagy preinitiation complex and that macrophages target Salmonella by Lc3-associated phagocytosis (LAP). In agreement, GFP-Lc3 recruitment and host resistance are impaired by deficiency of Rubcn/Rubicon, known as a negative regulator of canonical autophagy and an inducer of LAP. We also found strict dependency on NADPH oxidase, another essential factor for LAP. Both Rubcn and NADPH oxidase are required to activate a Salmonella biosensor for reactive oxygen species inside infected macrophages. These results identify LAP as the major host protective autophagy-related pathway responsible for macrophage defense against Salmonella during systemic infection. Abbreviations: ATG: autophagy related gene; BECN1: Beclin 1; CFU: colony forming units; CYBA/P22PHOX: cytochrome b-245, alpha chain; CYBB/NOX2: cytochrome b-245 beta chain; dpf: days post fertilization; EGFP: enhanced green fluorescent protein; GFP: green fluorescent protein; hfp: hours post fertilization; hpi: hours post infection; IRF8: interferon regulatory factor 8; Lcp1/L-plastin: lymphocyte cytosolic protein 1; LAP: LC3-associated phagocytosis; MAP1LC3/LC3: microtubule-associated protein 1A/1B-light chain 3; mCherry: red fluorescent protein; mpeg1: macrophage expressed gene 1; mpx: myeloid specific peroxidase; NADPH oxidase: nicotinamide adenine dinucleotide phosphate oxidase; NCF4/P40PHOX: neutrophil cytosolic factor 4; NTR-mCherry: nitroreductase-mCherry fusion; PTU: phenylthiourea; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol 3-phosphate; RB1CC1/FIP200: RB-1 inducible coiled coin 1; ROS: reactive oxygen species; RT-PCR: reverse transcriptase polymerase chain reaction; RUBCN/RUBICON: RUN and cysteine rich domain containing BECN1-interacting protein; SCV: Salmonella-containing vacuole; S. Typhimurium/S.T: Salmonella enterica serovar Typhimurium; TEM: transmission electron microscopy; Tg: transgenic; TSA: tyramide signal amplification; ULK1/2: unc-51-like autophagy activating kinase 1/2; UVRAG: UVRAG: UV radiation resistance associated; wt: wild type.


Subject(s)
Disease Models, Animal , Macrophages/physiology , Microtubule-Associated Proteins/physiology , Phagocytosis/genetics , Salmonella Infections, Animal , Salmonella typhimurium/immunology , Zebrafish Proteins/physiology , Zebrafish , Animals , Animals, Genetically Modified , Autophagy/physiology , Bacteremia/genetics , Bacteremia/immunology , Bacteremia/microbiology , Bacteremia/pathology , Embryo, Nonmammalian , Microtubule-Associated Proteins/genetics , Phagocytosis/immunology , Reactive Oxygen Species/metabolism , Salmonella Infections, Animal/genetics , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/metabolism , Salmonella Infections, Animal/microbiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/immunology , Zebrafish/microbiology , Zebrafish Proteins/genetics
12.
Development ; 145(4)2018 02 23.
Article in English | MEDLINE | ID: mdl-29475973

ABSTRACT

Resident microbes promote many aspects of host development, although the mechanisms by which microbiota influence host tissues remain unclear. We showed previously that the microbiota is required for allocation of appropriate numbers of secretory cells in the zebrafish intestinal epithelium. Because Notch signaling is crucial for secretory fate determination, we conducted epistasis experiments to establish whether the microbiota modulates host Notch signaling. We also investigated whether innate immune signaling transduces microbiota cues via the Myd88 adaptor protein. We provide the first evidence that microbiota-induced, Myd88-dependent signaling inhibits host Notch signaling in the intestinal epithelium, thereby promoting secretory cell fate determination. These results connect microbiota activity via innate immune signaling to the Notch pathway, which also plays crucial roles in intestinal homeostasis throughout life and when impaired can result in chronic inflammation and cancer.


Subject(s)
Intestinal Mucosa/metabolism , Microbiota , Myeloid Differentiation Factor 88/metabolism , Receptors, Notch/metabolism , Animals , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiology , Signal Transduction/physiology , Zebrafish/metabolism
14.
PLoS Pathog ; 13(6): e1006437, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28651010

ABSTRACT

Bacteria of the Burkholderia cepacia complex (Bcc) can cause devastating pulmonary infections in cystic fibrosis (CF) patients, yet the precise mechanisms underlying inflammation, recurrent exacerbations and transition from chronic stages to acute infection and septicemia are not known. Bcc bacteria are generally believed to have a predominant extracellular biofilm life style in infected CF lungs, similar to Pseudomonas aeruginosa, but this has been challenged by clinical observations which show Bcc bacteria predominantly in macrophages. More recently, Bcc bacteria have emerged in nosocomial infections of patients hospitalized for reasons unrelated to CF. Research has abundantly shown that Bcc bacteria can survive and replicate in mammalian cells in vitro, yet the importance of an intracellular life style during infection in humans is unknown. Here we studied the contribution of innate immune cell types to fatal pro-inflammatory infection caused by B. cenocepacia using zebrafish larvae. In strong contrast to the usual protective role for macrophages against microbes, our results show that these phagocytes significantly worsen disease outcome. We provide new insight that macrophages are critical for multiplication of B. cenocepacia in the host and for development of a fatal, pro-inflammatory response that partially depends on Il1-signalling. In contrast, neutrophils did not significantly contribute to disease outcome. In subcutaneous infections that are dominated by neutrophil-driven phagocytosis, the absence of a functional NADPH oxidase complex resulted in a small but measurably higher increase in bacterial growth suggesting the oxidative burst helps limit bacterial multiplication; however, neutrophils were unable to clear the bacteria. We suggest that paradigm-changing approaches are needed for development of novel antimicrobials to efficiently disarm intracellular bacteria of this group of highly persistent, opportunistic pathogens.


Subject(s)
Burkholderia cenocepacia/isolation & purification , Cross Infection/microbiology , Inflammation/microbiology , Macrophages/microbiology , Neutrophils/microbiology , Animals , Burkholderia Infections/immunology , Burkholderia cepacia complex/immunology , Cystic Fibrosis/complications , Humans , Lung/microbiology , Neutrophils/immunology , Phagocytosis/immunology , Pseudomonas aeruginosa/physiology , Respiratory Tract Infections/microbiology
15.
J Cereb Blood Flow Metab ; 35(1): 74-85, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25294126

ABSTRACT

Glucose transporter 2 (GLUT2; gene name SLC2A2) has a key role in the regulation of glucose dynamics in organs central to metabolism. Although GLUT2 has been studied in the context of its participation in peripheral and central glucose sensing, its role in the brain is not well understood. To decipher the role of GLUT2 in brain development, we knocked down slc2a2 (glut2), the functional ortholog of human GLUT2, in zebrafish. Abrogation of glut2 led to defective brain organogenesis, reduced glucose uptake and increased programmed cell death in the brain. Coinciding with the observed localization of glut2 expression in the zebrafish hindbrain, glut2 deficiency affected the development of neural progenitor cells expressing the proneural genes atoh1b and ptf1a but not those expressing neurod. Specificity of the morphant phenotype was demonstrated by the restoration of brain organogenesis, whole-embryo glucose uptake, brain apoptosis, and expression of proneural markers in rescue experiments. These results indicate that glut2 has an essential role during brain development by facilitating the uptake and availability of glucose and support the involvement of glut2 in brain glucose sensing.


Subject(s)
Brain/metabolism , Glucose Transporter Type 2/metabolism , Glucose/metabolism , Organogenesis/physiology , Zebrafish/embryology , Animals , Apoptosis/physiology , Brain/embryology , Brain/pathology , Cell Death , Cell Line , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Gene Knockdown Techniques , Glucose Transporter Type 2/genetics , Insulin-Secreting Cells/metabolism , Organogenesis/genetics , Real-Time Polymerase Chain Reaction , Transfection , Zebrafish/metabolism
16.
Autophagy ; 10(12): 2389-91, 2014.
Article in English | MEDLINE | ID: mdl-25484076

ABSTRACT

Autophagy provides an important defense mechanism against intracellular bacteria, such as Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis disease (TB). We recently reported that pathogen recognition and antibacterial autophagy are connected by the induction of the DNA damage-regulated autophagy modulator DRAM1 via the toll-like receptor (TLR)-MYD88-NFKB innate immunity signaling pathway. Having shown that DRAM1 colocalizes with Mtb in human macrophages, we took advantage of a zebrafish model for TB to investigate the function of DRAM1 in autophagic host defense in vivo. We found that DRAM1 protects the zebrafish host from infection with Mycobacterium marinum (Mm), a close relative of Mtb. Overexpression of DRAM1 increases autophagosome formation and promotes autophagic flux by a mechanism dependent on the cytosolic DNA sensor TMEM173/STING and the ubiquitin receptor SQSTM1/p62. Here we summarize and discuss the implications of these findings.


Subject(s)
Autophagy , Macrophages/microbiology , Membrane Proteins/metabolism , Mycobacterium Infections/metabolism , Mycobacterium/pathogenicity , Myeloid Differentiation Factor 88/metabolism , Animals , Humans
17.
J Virol ; 88(20): 12026-40, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25100833

ABSTRACT

Hemorrhagic viral diseases are distributed worldwide with important pathogens, such as dengue virus or hantaviruses. The lack of adequate in vivo infection models has limited the research on viral pathogenesis and the current understanding of the underlying infection mechanisms. Although hemorrhages have been associated with the infection of endothelial cells, other cellular types could be the main targets for hemorrhagic viruses. Our objective was to take advantage of the use of zebrafish larvae in the study of viral hemorrhagic diseases, focusing on the interaction between viruses and host cells. Cellular processes, such as transendothelial migration of leukocytes, virus-induced pyroptosis of macrophages. and interleukin-1ß (Il-1ß) release, could be observed in individual cells, providing a deeper knowledge of the immune mechanisms implicated in the disease. Furthermore, the application of these techniques to other pathogens will improve the current knowledge of host-pathogen interactions and increase the potential for the discovery of new therapeutic targets. Importance: Pathogenic mechanisms of hemorrhagic viruses are diverse, and most of the research regarding interactions between viruses and host cells has been performed in cell lines that might not be major targets during natural infections. Thus, viral pathogenesis research has been limited because of the lack of adequate in vivo infection models. The understanding of the relative pathogenic roles of the viral agent and the host response to the infection is crucial. This will be facilitated by the establishment of in vivo infection models using organisms such as zebrafish, which allows the study of the diseases in the context of a complete individual. The use of this animal model with other pathogens could improve the current knowledge on host-pathogen interactions and increase the potential for the discovery of new therapeutic targets against diverse viral diseases.


Subject(s)
Apoptosis , Interleukin-1beta/metabolism , Larva/metabolism , Macrophages/immunology , Zebrafish/growth & development , Animals , In Situ Nick-End Labeling , Larva/virology
18.
ACS Nano ; 8(7): 7014-26, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-24945994

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.


Subject(s)
Drug Carriers/chemistry , Embryo, Nonmammalian/microbiology , Mycobacterium marinum/drug effects , Nanoparticles/chemistry , Optical Imaging , Zebrafish/embryology , Zebrafish/microbiology , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biological Transport , Coumarins/chemistry , Drug Carriers/metabolism , Granuloma/microbiology , Lactic Acid/chemistry , Macrophages/microbiology , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/physiology , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rhodamines/chemistry , Rifampin/chemistry , Rifampin/pharmacology , Thiazoles/chemistry , Tuberculosis/microbiology , Tuberculosis/veterinary
19.
Cell Host Microbe ; 15(6): 753-67, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24922577

ABSTRACT

Autophagy is an important defense mechanism against mycobacteria, the causative agents of tuberculosis. The molecular mechanisms that link mycobacterial recognition to autophagy remain unclear. Our analysis in zebrafish and human macrophage models of mycobacterial infection reveals that the DNA damage-regulated autophagy modulator DRAM1 functions downstream of pathogen recognition by the Toll-like receptor (TLR)/interleukin-1 receptor (IL1R)-MYD88-NF-κB innate immune sensing pathway to activate selective autophagy. Mycobacterial infection of human macrophages and zebrafish embryos induced DRAM1 expression in a MYD88 and NF-κB-dependent manner. DRAM1 knockdown increased mycobacterial infection, whereas overexpression lowered infection by hyperactivating autophagy. DRAM1-mediated selective autophagic defenses require the cytosolic DNA sensor STING and the selective autophagy receptor p62/SQSTM1. Contrary to its known role in autophagy-mediated cell death and cancer, this DRAM1 function is p53 independent. We propose that DRAM1 mediates autophagic defense against a broader range of intracellular pathogens, since DRAM1 expression was also induced by the common bacterial endotoxin lipopolysaccharide.


Subject(s)
Autophagy , Macrophages/microbiology , Membrane Proteins/metabolism , Mycobacterium Infections/metabolism , Mycobacterium/pathogenicity , Myeloid Differentiation Factor 88/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy/immunology , Cells, Cultured , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/microbiology , Gene Expression Regulation , Genes, p53 , Host-Pathogen Interactions , Humans , Immunity, Innate , Lipopolysaccharides/pharmacology , Lysosomes/metabolism , Macrophages/physiology , Membrane Proteins/genetics , Mycobacterium Infections/immunology , Mycobacterium Infections/microbiology , NF-kappa B/metabolism , Receptors, Interleukin-1/metabolism , Sequestosome-1 Protein , Zebrafish/embryology , Zebrafish/microbiology
20.
PLoS One ; 9(6): e100928, 2014.
Article in English | MEDLINE | ID: mdl-24967596

ABSTRACT

Pulmonary tuberculosis (TB), caused by the intracellular bacterial pathogen Mycobacterium tuberculosis (Mtb), is a major world health problem. The production of reactive nitrogen species (RNS) is a potent cytostatic and cytotoxic defense mechanism against intracellular pathogens. Nevertheless, the protective role of RNS during Mtb infection remains controversial. Here we use an anti-nitrotyrosine antibody as a readout to study nitration output by the zebrafish host during early mycobacterial pathogenesis. We found that recognition of Mycobacterium marinum, a close relative of Mtb, was sufficient to induce a nitrosative defense mechanism in a manner dependent on MyD88, the central adaptor protein in Toll like receptor (TLR) mediated pathogen recognition. However, this host response was attenuated by mycobacteria via a virulence mechanism independent of the well-characterized RD1 virulence locus. Our results indicate a mechanism of pathogenic mycobacteria to circumvent host defense in vivo. Shifting the balance of host-pathogen interactions in favor of the host by targeting this virulence mechanism may help to alleviate the problem of infection with Mtb strains that are resistant to multiple drug treatments.


Subject(s)
Mycobacterium Infections/metabolism , Mycobacterium Infections/microbiology , Mycobacterium/physiology , Reactive Nitrogen Species/metabolism , Toll-Like Receptors/metabolism , Animals , Animals, Genetically Modified , Disease Models, Animal , Interleukin-8/metabolism , Mycobacterium Infections/immunology , Myeloid Differentiation Factor 88/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Peroxidase/metabolism , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-8B/metabolism , Signal Transduction , Tyrosine/metabolism , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...