Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Adv Healthc Mater ; 13(4): e2302596, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37935580

ABSTRACT

There is an urgent need for alternative antimicrobial materials due to the growing challenge of bacteria becoming resistant to conventional antibiotics. This study demonstrates the creation of a biocompatible pH-switchable antimicrobial material by combining bacteria-derived rhamnolipids (RL) and food-grade glycerol monooleate (GMO). The integration of RL into dispersed GMO particles, with an inverse-type liquid crystalline cubic structure in the core, leads to colloidally stable supramolecular materials. The composition and pH-triggered structural transformations are studied with small-angle X-ray scattering, cryogenic transmission electron microscopy, and dynamic light scattering. The composition-structure-activity relationship is analyzed and optimized to target bacteria at acidic pH values of acute wounds. The new RL/GMO dispersions reduce Staphylococcus aureus (S. aureus) populations by 7-log after 24 h of treatment with 64 µg mL-1 of RL and prevent biofilm formation at pH = 5.0, but have no activity at pH = 7.0. Additionally, the system is active against methicillin-resistant S. aureus (MRSA) with minimum inhibitory concentration of 128 µg mL-1 at pH 5.0. No activity is found against several Gram-negative bacteria at pH 5.0 and 7.0. The results provide a fundamental understanding of lipid self-assembly and the design of lipid-based biomaterials, which can further guide the development of alternative bio-based solutions to combat bacteria.


Subject(s)
Anti-Infective Agents , Methicillin-Resistant Staphylococcus aureus , Staphylococcus aureus , Glycolipids/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Infective Agents/pharmacology , Bacteria , Hydrogen-Ion Concentration , Microbial Sensitivity Tests
2.
Front Cell Dev Biol ; 10: 812244, 2022.
Article in English | MEDLINE | ID: mdl-35652104

ABSTRACT

Malaria affects the poorer regions of the world and is of tremendous health and economic burden for developing countries. Extracellular vesicles (EVs) are small vesicles released by almost any cells in the human body, including malaria infected red blood cells. Recent evidence shows that EVs might contribute to the pathogenesis of malaria. In addition, EVs hold considerable value in biomarker discovery. However, there are still significant gaps in our understanding of EV biology. So far most of our knowledge about EVs in malaria comes from in vitro work. More field studies are required to gain insight into their contribution to the disease and pathogenesis under physiological conditions. However, to perform research on EVs in low-income regions might be challenging due to the lack of appropriate equipment to isolate EVs. Therefore, there is a need to develop and validate EV extraction protocols applicable to poorly equipped laboratories. We established and validated two protocols for EV isolation from cell culture supernatants, rodent and human plasma. We compared polyethylene glycol (PEG) and salting out (SA) with sodium acetate for precipitation of EVs. We then characterized the EVs by Transmission Electron Microscopy (TEM), Western Blot, Size-exclusion chromatography (SEC), bead-based flow cytometry and protein quantification. Both protocols resulted in efficient purification of EVs without the need of expensive material or ultracentrifugation. Furthermore, the procedure is easily scalable to work with large and small sample volumes. Here, we propose that both of our approaches can be used in resource limited countries, therefore further helping to close the gap in knowledge of EVs during malaria.

3.
Front Immunol ; 13: 830290, 2022.
Article in English | MEDLINE | ID: mdl-35300343

ABSTRACT

Cytotoxic lymphocytes release proteins contained within the cytoplasmic cytolytic granules after recognition of infected or tumor target cells. These cytotoxic granular proteins (namely granzymes, granulysin, and perforin) are key immunological mediators within human cellular immunity. The availability of highly purified cytotoxic proteins has been fundamental for understanding their function in immunity and mechanistic involvement in sepsis and autoimmunity. Methods for recovery of native cytotoxic proteins can be problematic leading to: 1) the co-purification of additional proteins, confounding interpretation of function, and 2) low yields of highly purified proteins. Recombinant protein expression of individual cytolytic components can overcome these challenges. The use of mammalian expression systems is preferred for optimal post-translational modifications and avoidance of endotoxin contamination. Some of these proteins have been proposed for host directed human therapies (e.g. - granzyme A), or treatment of systemic infections or tumors as in granulysin. We report here a novel expression system using HEK293T cells for cost-effective purification of high yields of human granzymes (granzyme A and granzyme B) and granulysin with enhanced biological activity than previous reports. The resulting proteins are free of native contaminants, fold correctly, and remain enzymatically active. Importantly, these improvements have also led to the first purification of biologically active recombinant human granulysin in high yields from a mammalian system. This method can be used as a template for purification of many other secreted cellular proteins and may lead to advances for human medicine.


Subject(s)
Mammals , Animals , Cytoplasm/metabolism , Granzymes/metabolism , HEK293 Cells , Humans , Mammals/metabolism , Perforin
4.
Front Immunol ; 12: 755856, 2021.
Article in English | MEDLINE | ID: mdl-34899706

ABSTRACT

The immune system protects the host from a plethora of microorganisms and toxins through its unique ability to distinguish self from non-self. To perform this delicate but essential task, the immune system relies on two lines of defense. The innate immune system, which is by nature fast acting, represents the first line of defense. It involves anatomical barriers, physiological factors as well as a subset of haematopoietically-derived cells generically call leukocytes. Activation of the innate immune response leads to a state of inflammation that serves to both warn about and combat the ongoing infection and delivers the antigenic information of the invading pathogens to initiate the slower but highly potent and specific second line of defense, the adaptive immune system. The adaptive immune response calls on T lymphocytes as well as the B lymphocytes essential for the elimination of pathogens and the establishment of the immunological memory. Reactive oxygen species (ROS) have been implicated in many aspects of the immune responses to pathogens, mostly in innate immune functions, such as the respiratory burst and inflammasome activation. Here in this mini review, we focus on the role of ROS in adaptive immunity. We examine how ROS contribute to T-cell biology and discuss whether this activity can be extrapolated to B cells.


Subject(s)
Adaptive Immunity/immunology , Reactive Oxygen Species/immunology , Animals , B-Lymphocytes/immunology , Humans , T-Lymphocytes/immunology
5.
Front Immunol ; 12: 750512, 2021.
Article in English | MEDLINE | ID: mdl-34707614

ABSTRACT

Cell-mediated cytotoxicity is an essential immune defense mechanism to fight against viral, bacterial or parasitic infections. Upon recognition of an infected target cell, killer lymphocytes form an immunological synapse to release the content of their cytotoxic granules. Cytotoxic granules of humans contain two membrane-disrupting proteins, perforin and granulysin, as well as a homologous family of five death-inducing serine proteases, the granzymes. The granzymes, after delivery into infected host cells by the membrane disrupting proteins, may contribute to the clearance of microbial pathogens through different mechanisms. The granzymes can induce host cell apoptosis, which deprives intracellular pathogens of their protective niche, therefore limiting their replication. However, many obligate intracellular pathogens have evolved mechanisms to inhibit programed cells death. To overcome these limitations, the granzymes can exert non-cytolytic antimicrobial activities by directly degrading microbial substrates or hijacked host proteins crucial for the replication or survival of the pathogens. The granzymes may also attack factors that mediate microbial virulence, therefore directly affecting their pathogenicity. Many mechanisms applied by the granzymes to eliminate infected cells and microbial pathogens rely on the induction of reactive oxygen species. These reactive oxygen species may be directly cytotoxic or enhance death programs triggered by the granzymes. Here, in the light of the latest advances, we review the antimicrobial activities of the granzymes in regards to their cytolytic and non-cytolytic activities to inhibit pathogen replication and invasion. We also discuss how reactive oxygen species contribute to the various antimicrobial mechanisms exerted by the granzymes.


Subject(s)
Granzymes/immunology , Animals , Cell Death , Humans , Infections/immunology , Reactive Oxygen Species/immunology
6.
Front Immunol ; 12: 712678, 2021.
Article in English | MEDLINE | ID: mdl-34413857

ABSTRACT

Mycobacterium tuberculosis (Mtb), the pathological agent that causes tuberculosis (TB) is the number one infectious killer worldwide with one fourth of the world's population currently infected. Data indicate that γ9δ2 T cells secrete Granzyme A (GzmA) in the extracellular space triggering the infected monocyte to inhibit growth of intracellular mycobacteria. Accordingly, deletion of GZMA from γ9δ2 T cells reverses their inhibitory capacity. Through mechanistic studies, GzmA's action was investigated in monocytes from human PBMCs. The use of recombinant human GzmA expressed in a mammalian system induced inhibition of intracellular mycobacteria to the same degree as previous human native protein findings. Our data indicate that: 1) GzmA is internalized within mycobacteria-infected cells, suggesting that GzmA uptake could prevent infection and 2) that the active site is not required to inhibit intracellular replication. Global proteomic analysis demonstrated that the ER stress response and ATP producing proteins were upregulated after GzmA treatment, and these proteins abundancies were confirmed by examining their expression in an independent set of patient samples. Our data suggest that immunotherapeutic host interventions of these pathways may contribute to better control of the current TB epidemic.


Subject(s)
Adenosine Triphosphate/biosynthesis , Endoplasmic Reticulum Stress/immunology , Granzymes/physiology , Monocytes/microbiology , Mycobacterium bovis/physiology , T-Lymphocyte Subsets/immunology , Blotting, Western , Cell Division , Granzymes/biosynthesis , Granzymes/genetics , Granzymes/pharmacology , HEK293 Cells , Humans , Memory T Cells/immunology , Memory T Cells/metabolism , Proteome , Receptors, Antigen, T-Cell, gamma-delta/analysis , Recombinant Proteins/pharmacology , T-Lymphocyte Subsets/metabolism , Two-Dimensional Difference Gel Electrophoresis
7.
Front Immunol ; 12: 643746, 2021.
Article in English | MEDLINE | ID: mdl-34093532

ABSTRACT

Malaria remains one of the most serious health problems in developing countries. The causative agent of malaria, Plasmodium spp., have a complex life cycle involving multiple developmental stages as well as different morphological, biochemical and metabolic requirements. We recently found that γδ T cells control parasite growth using pore-forming proteins to deliver their cytotoxic proteases, the granzymes, into blood residing parasites. Here, we follow up on the molecular mechanisms of parasite growth inhibition by human pore-forming proteins. We confirm that Plasmodium falciparum infection efficiently depletes the red blood cells of cholesterol, which renders the parasite surrounding membranes susceptible to lysis by prokaryotic membrane disrupting proteins, such as lymphocytic granulysin or the human cathelicidin LL-37. Interestingly, not the cholesterol depletion but rather the simultaneous exposure of phosphatidylserine, a negatively charged phospholipid, triggers resistance of late stage parasitized red blood cells towards the eukaryotic pore forming protein perforin. Overall, by revealing the molecular events we establish here a pathogen-host interaction that involves host cell membrane remodeling that defines the susceptibility towards cytolytic molecules.


Subject(s)
Erythrocyte Membrane/immunology , Hemolysis/immunology , Malaria, Falciparum/immunology , Perforin/immunology , Plasmodium falciparum/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Antigens, Differentiation, T-Lymphocyte , Antimicrobial Cationic Peptides/immunology , Disease Susceptibility , Erythrocyte Membrane/parasitology , Humans , Cathelicidins
9.
iScience ; 23(3): 100932, 2020 Mar 27.
Article in English | MEDLINE | ID: mdl-32151975

ABSTRACT

Pathogenic bacteria secrete virulence factors that interact with the human host to establish infections. The human immune system evolved multiple mechanisms to fight bacterial invaders, including immune proteases that were demonstrated to contribute crucially to antibacterial defense. Here we show that granzyme B degrades multiple secreted virulence mediators from Listeria monocytogenes, Salmonella typhimurium, and Mycobacteria tuberculosis. Pathogenic bacteria, when infected in the presence of granzyme B or granzyme-secreting killer cells, fail to grow in human macrophages and epithelial cells owing to their crippled virulence. A granzyme B-uncleavable mutant form of the major Listeria virulence factor, listeriolysin O, rescued the virulence defect in response to granzyme treatment. Hence, we link the degradation of a single factor with the observed decrease in virulent bacteria growth. Overall, we reveal here an innate immune barrier function of granzyme B by disrupting bacterial virulence to facilitate bacteria clearance by bystander immune and non-immune cells.

10.
Front Immunol ; 11: 22, 2020.
Article in English | MEDLINE | ID: mdl-32082312

ABSTRACT

Malaria infection caused by the Plasmodium species is a complex disease in which a fine balance between host and parasite factors determine the disease severity. While in some individuals, the infection will trigger only a mild and uncomplicated disease, other individuals will develop severe complications which lead to death. Extracellular vesicles (EVs) secreted by infected red blood cells (iRBCs), as well as other host cells, are important regulators of the balance that determines the disease outcome. In addition, EVs constitute a robust mode of cell-to-cell communication by transferring signaling cargoes between parasites, and between parasites and host, without requiring cellular contact. The transfer of membrane and cytosolic proteins, lipids, DNA, and RNA through EVs not only modulate the immune response, it also mediates cellular communication between parasites to synchronize the transmission stage. Here, we review the recent progress in understanding EV roles during malaria.


Subject(s)
Cell Communication/immunology , Extracellular Vesicles/metabolism , Malaria/immunology , Plasmodium/growth & development , Signal Transduction/immunology , Animals , Disease Models, Animal , Erythrocytes/immunology , Erythrocytes/metabolism , Erythrocytes/parasitology , Extracellular Vesicles/parasitology , Host-Parasite Interactions/immunology , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/parasitology , Life Cycle Stages , Malaria/parasitology , Mice , RNA/metabolism
11.
J Immunol ; 204(7): 1798-1809, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32066596

ABSTRACT

Plasmodium spp., the causative agent of malaria, have a complex life cycle. The exponential growth of the parasites during the blood stage is responsible for almost all malaria-associated morbidity and mortality. Therefore, tight immune control of the intraerythrocytic replication of the parasite is essential to prevent clinical malaria. Despite evidence that the particular lymphocyte subset of γδ T cells contributes to protective immunity during the blood stage in naive hosts, their precise inhibitory mechanisms remain unclear. Using human PBMCs, we confirmed in this study that γδ T cells specifically and massively expanded upon activation with Plasmodium falciparum culture supernatant. We also demonstrate that these activated cells gain cytolytic potential by upregulating cytotoxic effector proteins and IFN-γ. The killer cells bound to infected RBCs and killed intracellular P. falciparum via the transfer of the granzymes, which was mediated by granulysin in a stage-specific manner. Several vital plasmodial proteins were efficiently destroyed by granzyme B, suggesting proteolytic degradation of these proteins as essential in the lymphocyte-mediated death pathway. Overall, these data establish a granzyme- and granulysin-mediated innate immune mechanism exerted by γδ T cells to kill late-stage blood-residing P. falciparum.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/immunology , Granzymes/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Antigens, Protozoan/immunology , Cells, Cultured , Erythrocytes/immunology , Humans , Immunity, Innate/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/immunology , Life Cycle Stages/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Up-Regulation/immunology
12.
Pathogens ; 9(1)2019 Dec 24.
Article in English | MEDLINE | ID: mdl-31878288

ABSTRACT

Microglia are the chief immune cells of the brain and have been reported to be activated in severe malaria. Their activation may drive towards neuroinflammation in cerebral malaria. Malaria-infected red blood cell derived-extracellular vesicles (MiREVs) are produced during the blood stage of malaria infection. They mediate intercellular communication and immune regulation, among other functions. During cerebral malaria, the breakdown of the blood-brain barrier can promote the migration of substances such as MiREVs from the periphery into the brain, targeting cells such as microglia. Microglia and extracellular vesicle interactions in different pathological conditions have been reported to induce neuroinflammation. Unlike in astrocytes, microglia-extracellular vesicle interaction has not yet been described in malaria infection. Therefore, in this study, we aimed to investigate the uptake of MiREVs by human microglia cells and their cytokine response. Human blood monocyte-derived microglia (MoMi) were generated from buffy coats of anonymous healthy donors using Ficoll-Paque density gradient centrifugation. The MiREVs were isolated from the Plasmodium falciparum cultures. They were purified by ultracentrifugation and labeled with PKH67 green fluorescent dye. The internalization of MiREVs by MoMi was observed after 4 h of co-incubation on coverslips placed in a 24-well plate at 37 °C using confocal microscopy. Cytokine-gene expression was investigated using rt-qPCR, following the stimulation of the MoMi cells with supernatants from the parasite cultures at 2, 4, and 24 h, respectively. MiREVs were internalized by the microglia and accumulated in the perinuclear region. MiREVs-treated cells increased gene expression of the inflammatory cytokine TNFα and reduced gene expression of the immune suppressive IL-10. Overall, the results indicate that MiREVs may act on microglia, which would contribute to enhanced inflammation in cerebral malaria.

13.
J Vis Exp ; (132)2018 02 14.
Article in English | MEDLINE | ID: mdl-29553510

ABSTRACT

Malaria is a life-threatening disease caused by Plasmodium parasites, with P. falciparum being the most prevalent on the African continent and responsible for most malaria-related deaths globally. Several factors including parasite sequestration in tissues, vascular dysfunction, and inflammatory responses influence the evolution of the disease in malaria-infected people. P. falciparum-infected red blood cells (iRBCs) release small extracellular vesicles (EVs) containing different kinds of cargo molecules that mediate pathogenesis and cellular communication between parasites and host. EVs are efficiently taken up by cells in which they modulate their function. Here we discuss strategies to address the role of EVs in parasite-host interactions. First, we describe a straightforward method for labeling and tracking EV internalization by endothelial cells, using a green cell linker dye. Second, we report a simple way to measure permeability across an endothelial cell monolayer by using a fluorescently labeled dextran. Finally, we show how to investigate the role of small non-coding RNA molecules in endothelial cell function.


Subject(s)
Endothelial Cells/pathology , Erythrocytes/pathology , Erythrocytes/parasitology , Extracellular Vesicles/pathology , Malaria, Falciparum/blood , Animals , Endothelial Cells/metabolism , Erythrocytes/metabolism , Extracellular Vesicles/metabolism , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Microscopy, Confocal
14.
Sci Rep ; 8(1): 884, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29343745

ABSTRACT

The parasite Plasmodium falciparum causes the most severe form of malaria. Cell communication between parasites is an important mechanism to control population density and differentiation. The infected red blood cells (iRBCs) release small extracellular vesicles (EVs) that transfer cargoes between cells. The EVs synchronize the differentiation of the asexual parasites into gametocytes to initiate the transmission to the mosquito. Beside their role in parasite communication, EVs regulate vascular function. So far, the exact cargoes responsible for cellular communication remain unknown. We isolated EVs from cultured iRBCs to determine their small RNA content. We identified several types of human and plasmodial regulatory RNAs. While the miRNAs and tRNA-derived fragments were the most abundant human RNAs, we also found Y-RNAs, vault RNAs, snoRNAs and piRNAs. Interestingly, we found about 120 plasmodial RNAs, including mRNAs coding for exported proteins and proteins involved in drug resistance, as well as non-coding RNAs, such as rRNAs, small nuclear (snRNAs) and tRNAs. These data show, that iRBC-EVs carry small regulatory RNAs. A role in cellular communication is possible since the RNAs were transferred to endothelial cells. Furthermore, the presence of Plasmodium RNAs, in EVs suggests that they may be used as biomarker to track and detect disease.


Subject(s)
Erythrocytes/parasitology , Extracellular Vesicles/genetics , Malaria/genetics , RNA/genetics , Cell Communication/genetics , Cell Differentiation/genetics , Cells, Cultured , Endothelial Cells/parasitology , Erythrocyte Count/methods , Extracellular Vesicles/parasitology , Humans , Malaria/parasitology , Plasmodium falciparum/pathogenicity
15.
Nanomedicine ; 14(2): 601-607, 2018 02.
Article in English | MEDLINE | ID: mdl-29155361

ABSTRACT

Bone infections are difficult to treat and can lead to severe tissue destruction. Acute bone infections are usually caused by Staphylococcus aureus. Osteoclasts, which belong to the monocyte/macrophage lineage, are the key cells in bone infections. They are not well equipped for killing bacteria and may serve as a reservoir for bacterial pathogens. Silver has been known for centuries for its bactericidal activity. Here, we investigated the bactericidal effects of nano-silver particles in bacteria infected human osteoclasts. We found that nano-silver in per se non-toxic concentration enhanced the bactericidal activity in osteoclasts against intracellular Methicillin-resistant, virulent Staphylococcus aureus. The reduced bacterial survival in nano-silver pretreated cells correlated with increased reactive oxygen responses towards the invading pathogens. Overall, these results indicate that nano-silver compounds should be considered as an effective treatment and prevention option for bacterial bone and orthopedic implant infections.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Metal Nanoparticles/administration & dosage , Osteoclasts/drug effects , Reactive Oxygen Species/metabolism , Silver/chemistry , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/chemistry , Cells, Cultured , Humans , Metal Nanoparticles/chemistry , Osteoclasts/pathology , Phagocytosis , Staphylococcal Infections/microbiology , Staphylococcus aureus/isolation & purification
16.
Methods Mol Biol ; 1660: 377-388, 2017.
Article in English | MEDLINE | ID: mdl-28828673

ABSTRACT

Growing attention is drawn toward the role of extracellular vesicles (EVs) in infectious diseases. EVs, which are small vesicles released by cells, are involved in cellular communication, immune regulation, and pathogenesis. EVs act as messenger carrying functional cargoes, including RNA, DNA, lipids and proteins from a donor cell to regulate the function of a recipient cell. In malaria, EVs play a key role in regulating the progression from the blood to the transmission stage by promoting the switch between asexual and sexual stages that are taken up by mosquitoes. In addition to their role in parasite communication, EVs modulate the immune system and regulate endothelial cell function.In this chapter, we describe protocols to isolate, purify and characterize EVs derived from Plasmodium falciparum infected red blood cell culture.


Subject(s)
Erythrocytes/metabolism , Erythrocytes/parasitology , Malaria/metabolism , Malaria/parasitology , Cell Communication , Chromatography, Gel , Chromatography, Liquid , Extracellular Vesicles/metabolism , Humans , Plasmodium falciparum , Ultracentrifugation
17.
Cell Death Differ ; 24(4): 747-758, 2017 04.
Article in English | MEDLINE | ID: mdl-28338658

ABSTRACT

We have found that granzyme B (GB)-induced apoptosis also requires reactive oxygen species resulting from the alteration of mitochondrial complex I. How GB, which does not possess a mitochondrial targeting sequence, enter this organelle is unknown. We show that GB enters the mitochondria independently of the translocase of the outer mitochondrial membrane complex, but requires instead Sam50, the central subunit of the sorting and assembly machinery that integrates outer membrane ß-barrel proteins. Moreover, GB breaches the inner membrane through Tim22, the metabolite carrier translocase pore, in a mitochondrial heat-shock protein 70 (mtHsp70)-dependent manner. Granzyme A (GA) and caspase-3 use a similar route to the mitochondria. Finally, preventing GB from entering the mitochondria either by mutating lysine 243 and arginine 244 or depleting Sam50 renders cells more resistant to GB-mediated reactive oxygen species and cell death. Similarly, Sam50 depletion protects cells from GA-, GM- and caspase-3-mediated cell death. Therefore, cytotoxic molecules enter the mitochondria to induce efficiently cell death through a noncanonical Sam50-, Tim22- and mtHsp70-dependent import pathway.


Subject(s)
Apoptosis , Granzymes/metabolism , HSP70 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Doxorubicin/toxicity , Electron Transport Complex I/metabolism , Granzymes/antagonists & inhibitors , Granzymes/genetics , HeLa Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondrial Membranes/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Valinomycin/toxicity
18.
Virol J ; 14(1): 8, 2017 01 14.
Article in English | MEDLINE | ID: mdl-28088249

ABSTRACT

BACKGROUND: Japanese encephalitis virus (JEV) is a neurotropic flavivirus causing mortality and morbidity in humans. Severe Japanese encephalitis cases display strong inflammatory responses in the central nervous system and an accumulation of viral particles in specific brain regions. Microglia cells are the unique brain-resident immune cell population with potent migratory functions and have been proposed to act as a viral reservoir for JEV. Animal models suggest that the targeting of microglia by JEV is partially responsible for inflammatory reactions in the brain. Nevertheless, the interactions between human microglia and JEV are poorly documented. METHODS: Using human primary microglia and a new model of human blood monocyte-derived microglia, the present study explores the interaction between human microglia and JEV as well as the role of these cells in viral transmission to susceptible cells. To achieve this work, vaccine-containing inactivated JEV and two live JEV strains were applied on human microglia. RESULTS: Live JEV was non-cytopathogenic to human microglia but increased levels of CCL2, CXCL9 and CXCL10 in such cultures. Furthermore, human microglia up-regulated the expression of the fraktalkine receptor CX3CR1 upon exposure to both JEV vaccine and live JEV. Although JEV vaccine enhanced MHC class II on all microglia, live JEV enhanced MHC class II mainly on CX3CR1+ microglia cells. Importantly, human microglia supported JEV replication, but infectivity was only transmitted to neighbouring cells in a contact-dependent manner. CONCLUSION: Our findings suggest that human microglia may be a source of neuronal infection and sustain JEV brain pathogenesis.


Subject(s)
Encephalitis Virus, Japanese/physiology , Host-Pathogen Interactions , Microglia/virology , Virus Replication , Cells, Cultured , Chemokines/biosynthesis , Humans
19.
Methods Mol Biol ; 1535: 275-284, 2017.
Article in English | MEDLINE | ID: mdl-27914086

ABSTRACT

Bacterial pathogens represent a constant threat to human health that was exacerbated in recent years by a dramatic increase of strains resistant to last resort antibiotics. The immune system of higher vertebrates generally evolved several efficient innate and adaptive mechanisms to fight ubiquitous bacterial pathogens. Among those mechanisms, immune proteases were recognized to contribute essentially to antibacterial immune defense. The effector serine proteases of the adaptive immune system, the granzymes, exert potent antimicrobial activity when they are delivered into the bacterial cytosol by prokaryotic membrane disrupting proteins, such as granulysin.In this chapter, we are detailing experimental protocols to study the synergistic cytotoxic effects of human granzymes and granulysin on extracellular as well as on intracellular bacterial pathogens in vitro. In addition, we provide a simple and fast-forward method to biochemically purify native cytotoxic effector molecules necessary to perform this kind of investigations.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antigens, Differentiation, T-Lymphocyte/pharmacology , Bacteria/drug effects , Bacteria/immunology , Cytotoxicity, Immunologic , Granzymes/pharmacology , Perforin/pharmacology , Cell Line , Dose-Response Relationship, Drug , Extracellular Space/immunology , Extracellular Space/microbiology , Humans , Intracellular Space/immunology , Intracellular Space/microbiology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
20.
Nat Commun ; 7: 12727, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27721445

ABSTRACT

Malaria remains one of the greatest public health challenges worldwide, particularly in sub-Saharan Africa. The clinical outcome of individuals infected with Plasmodium falciparum parasites depends on many factors including host systemic inflammatory responses, parasite sequestration in tissues and vascular dysfunction. Production of pro-inflammatory cytokines and chemokines promotes endothelial activation as well as recruitment and infiltration of inflammatory cells, which in turn triggers further endothelial cell activation and parasite sequestration. Inflammatory responses are triggered in part by bioactive parasite products such as hemozoin and infected red blood cell-derived extracellular vesicles (iRBC-derived EVs). Here we demonstrate that such EVs contain functional miRNA-Argonaute 2 complexes that are derived from the host RBC. Moreover, we show that EVs are efficiently internalized by endothelial cells, where the miRNA-Argonaute 2 complexes modulate target gene expression and barrier properties. Altogether, these findings provide a mechanistic link between EVs and vascular dysfunction during malaria infection.


Subject(s)
Argonaute Proteins/metabolism , Blood Vessels/metabolism , Erythrocytes/parasitology , Extracellular Vesicles/metabolism , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , MicroRNAs/metabolism , Brain/blood supply , Cell Line , Endocytosis , Endothelial Cells/metabolism , Erythrocytes/ultrastructure , Extracellular Vesicles/ultrastructure , Gene Expression Regulation , Gene Silencing , Humans , MicroRNAs/genetics , Microvessels/cytology , RNA-Induced Silencing Complex/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...