Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biomedicines ; 12(2)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38398037

ABSTRACT

Proteolysis of the extracellular matrix (ECM) by matrix metalloproteinases (MMPs) plays a crucial role in the immune response to bacterial infections. Here we report the secretion of MMPs associated with proteolytic extracellular vesicles (EVs) released by macrophages in response to Salmonella enterica serovar Typhimurium infection. Specifically, we used global proteomics, in vitro, and in vivo approaches to investigate the composition and function of these proteolytic EVs. Using a model of S. Typhimurium infection in murine macrophages, we isolated and characterized a population of small EVs. Bulk proteomics analysis revealed significant changes in protein cargo of naïve and S. Typhimurium-infected macrophage-derived EVs, including the upregulation of MMP-9. The increased levels of MMP-9 observed in immune cells exposed to S. Typhimurium were found to be regulated by the toll-like receptor 4 (TLR-4)-mediated response to bacterial lipopolysaccharide. Macrophage-derived EV-associated MMP-9 enhanced the macrophage invasion through Matrigel as selective inhibition of MMP-9 reduced macrophage invasion. Systemic administration of fluorescently labeled EVs into immunocompromised mice demonstrated that EV-associated MMP activity facilitated increased accumulation of EVs in spleen and liver tissues. This study suggests that macrophages secrete proteolytic EVs to enhance invasion and ECM remodeling during bacterial infections, shedding light on an essential aspect of the immune response.

2.
ACS Nano ; 16(8): 12276-12289, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35921522

ABSTRACT

The elucidation of viral-receptor interactions and an understanding of virus-spreading mechanisms are of great importance, particularly in the era of a pandemic. Indeed, advances in computational chemistry, synthetic biology, and protein engineering have allowed precise prediction and characterization of such interactions. Nevertheless, the hazards of the infectiousness of viruses, their rapid mutagenesis, and the need to study viral-receptor interactions in a complex in vivo setup call for further developments. Here, we show the development of biocompatible genetically engineered extracellular vesicles (EVs) that display the receptor binding domain (RBD) of SARS-CoV-2 on their surface as coronavirus mimetics (EVsRBD). Loading EVsRBD with iron oxide nanoparticles makes them MRI-visible and, thus, allows mapping of the binding of RBD to ACE2 receptors noninvasively in live subjects. Moreover, we show that EVsRBD can be modified to display mutants of the RBD of SARS-CoV-2, allowing rapid screening of currently raised or predicted variants of the virus. The proposed platform thus shows relevance and cruciality in the examination of quickly evolving pathogenic viruses in an adjustable, fast, and safe manner. Relying on MRI for visualization, the presented approach could be considered in the future to map ligand-receptor binding events in deep tissues, which are not accessible to luminescence-based imaging.


Subject(s)
COVID-19 , Extracellular Vesicles , Humans , SARS-CoV-2/genetics , Angiotensin-Converting Enzyme 2 , Spike Glycoprotein, Coronavirus/chemistry , Peptidyl-Dipeptidase A/metabolism , Binding Sites , Protein Binding , Extracellular Vesicles/metabolism , Magnetic Resonance Imaging
3.
Methods Mol Biol ; 2470: 133-145, 2022.
Article in English | MEDLINE | ID: mdl-35881344

ABSTRACT

Malaria is one the most devastating infectious diseases in the world: of the five malaria-associated parasites, Plasmodium falciparum and P. vivax are the most pathogenic and widespread, respectively. P. falciparum invades human red blood cells (RBCs), releasing extracellular vesicles (Pf-EV) carrying DNA, RNA and protein cargo components involved in host-pathogen communications in the course of the disease. Different strategies have been used to analyze Pf-EV biophysically and chemically. Atomic force microscopy (AFM) stands out as a powerful tool for rendering high quality images of extracellular vesicles. In this technique, a sharp tip attached to a cantilever reconstructs the topographic surface of the extracellular vesicles and probes their nano-mechanical properties based on force-distance curves. Here, we describe a method to separate Pf-EV using differential ultracentrifugation, followed by nanoparticle tracking analysis (NTA) to quantify and estimate the size distribution. Finally, the AFM imaging procedure on Pf-EV adsorbed on a Mg2+-modified mica surface is detailed.


Subject(s)
Extracellular Vesicles , Malaria, Falciparum , Malaria , Erythrocytes/metabolism , Extracellular Vesicles/metabolism , Humans , Malaria/parasitology , Malaria, Falciparum/parasitology , Microscopy, Atomic Force , Plasmodium falciparum , Plasmodium vivax
4.
EMBO Rep ; 23(7): e54755, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35642585

ABSTRACT

Malaria is the most serious mosquito-borne parasitic disease, caused mainly by the intracellular parasite Plasmodium falciparum. The parasite invades human red blood cells and releases extracellular vesicles (EVs) to alter its host responses. It becomes clear that EVs are generally composed of sub-populations. Seeking to identify EV subpopulations, we subject malaria-derived EVs to size-separation analysis, using asymmetric flow field-flow fractionation. Multi-technique analysis reveals surprising characteristics: we identify two distinct EV subpopulations differing in size and protein content. Small EVs are enriched in complement-system proteins and large EVs in proteasome subunits. We then measure the membrane fusion abilities of each subpopulation with three types of host cellular membranes: plasma, late and early endosome. Remarkably, small EVs fuse to early endosome liposomes at significantly greater levels than large EVs. Atomic force microscope imaging combined with machine-learning methods further emphasizes the difference in biophysical properties between the two subpopulations. These results shed light on the sophisticated mechanism by which malaria parasites utilize EV subpopulations as a communication tool to target different cellular destinations or host systems.


Subject(s)
Extracellular Vesicles , Malaria , Parasites , Animals , Erythrocytes/parasitology , Extracellular Vesicles/metabolism , Humans , Plasmodium falciparum
5.
Immunity ; 55(3): 442-458.e8, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35182483

ABSTRACT

Consecutive exposures to different pathogens are highly prevalent and often alter the host immune response. However, it remains unknown how a secondary bacterial infection affects an ongoing adaptive immune response elicited against primary invading pathogens. We demonstrated that recruitment of Sca-1+ monocytes into lymphoid organs during Salmonella Typhimurium (STm) infection disrupted pre-existing germinal center (GC) reactions. GC responses induced by influenza, plasmodium, or commensals deteriorated following STm infection. GC disruption was independent of the direct bacterial interactions with B cells and instead was induced through recruitment of CCR2-dependent Sca-1+ monocytes into the lymphoid organs. GC collapse was associated with impaired cellular respiration and was dependent on TNFα and IFNγ, the latter of which was essential for Sca-1+ monocyte differentiation. Monocyte recruitment and GC disruption also occurred during LPS-supplemented vaccination and Listeria monocytogenes infection. Thus, systemic activation of the innate immune response upon severe bacterial infection is induced at the expense of antibody-mediated immunity.


Subject(s)
Bacterial Infections , Listeriosis , B-Lymphocytes , Germinal Center , Humans , Monocytes
6.
PNAS Nexus ; 1(4): pgac156, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36714848

ABSTRACT

Extracellular vesicles (EVs) transfer bioactive molecules between cells in a process reminiscent of enveloped viruses. EV cargo delivery is thought to occur by protein-mediated and pH-dependent membrane fusion of the EV and the cellular membrane. However, there is a lack of methods to identify the fusion proteins and resolve their mechanism. We developed and benchmarked an in vitro biophysical assay to investigate EV membrane fusion. The assay was standardized by directly comparing EV and viral fusion with liposomes. We show that EVs and retroviruses fuse with liposomes mimicking the membrane composition of the late endosome in a pH- and protein-dependent manner. Moreover, we directly visualize the stages of membrane fusion using cryo-electron tomography. We find that, unlike most retroviruses, EVs remain fusogenic after acidification and reneutralization. These results provide novel insights into the EV cargo delivery mechanism and an experimental approach to identify the EV fusion machinery.

7.
Nat Commun ; 12(1): 4851, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34381047

ABSTRACT

Pathogens are thought to use host molecular cues to control when to initiate life-cycle transitions, but these signals are mostly unknown, particularly for the parasitic disease malaria caused by Plasmodium falciparum. The chemokine CXCL10 is present at high levels in fatal cases of cerebral malaria patients, but is reduced in patients who survive and do not have complications. Here we show a Pf 'decision-sensing-system' controlled by CXCL10 concentration. High CXCL10 expression prompts P. falciparum to initiate a survival strategy via growth acceleration. Remarkably, P. falciparum inhibits CXCL10 synthesis in monocytes by disrupting the association of host ribosomes with CXCL10 transcripts. The underlying inhibition cascade involves RNA cargo delivery into monocytes that triggers RIG-I, which leads to HUR1 binding to an AU-rich domain of the CXCL10 3'UTR. These data indicate that when the parasite can no longer keep CXCL10 at low levels, it can exploit the chemokine as a cue to shift tactics and escape.


Subject(s)
Chemokine CXCL10/metabolism , Malaria, Falciparum/parasitology , Plasmodium falciparum/physiology , 3' Untranslated Regions , Chemokine CXCL10/genetics , DEAD Box Protein 58/metabolism , ELAV-Like Protein 1/metabolism , Extracellular Vesicles/metabolism , Host-Parasite Interactions , Humans , Life Cycle Stages , Malaria, Falciparum/immunology , Monocytes/metabolism , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Protein Biosynthesis , RNA, Protozoan/metabolism , Receptors, Immunologic/metabolism , Ribosomes/metabolism , THP-1 Cells
8.
Front Cell Infect Microbiol ; 11: 739628, 2021.
Article in English | MEDLINE | ID: mdl-35155269

ABSTRACT

Extracellular vesicles (EVs) are produced by across almost all the living kingdoms and play a crucial role in cell-cell communication processes. EVs are especially important for pathogens, as Plasmodium falciparum (Pf) parasite, the leading causing species in human malaria. Malaria parasites are able to modulate the host immune response from a distance via delivering diverse cargo components inside the EVs, such as proteins and nucleic acids. We have previously shown that imaging flow cytometry (IFC) can be effectively used to monitor the uptake of different cargo components of malaria derived EVs by host human monocytes. Here, we take this approach one step further and demonstrate that we can directly investigate the dynamics of the cargo distribution pattern over time by monitoring its distribution within two different recipient cells of the immune system, monocytes vs macrophages. By staining the RNA cargo of the vesicles and monitor the signal we were able to evaluate the kinetics of its delivery and measure different parameters of the cargo's distribution post internalization. Interestingly, we found that while the level of the EV uptake is similar, the pattern of the signal for RNA cargo distribution is significantly different between these two recipient immune cells. Our results demonstrate that this method can be applied to study the distribution dynamics of the vesicle cargo post uptake to different types of cells. This can benefit significantly to our understanding of the fate of cargo components post vesicle internalization in the complex interface between pathogen-derived vesicles and their host recipient cells.


Subject(s)
Extracellular Vesicles , Monocytes , Cell Communication/genetics , Extracellular Vesicles/metabolism , Humans , Macrophages , Monocytes/metabolism , RNA/metabolism
9.
Biomedicines ; 8(5)2020 Apr 27.
Article in English | MEDLINE | ID: mdl-32349226

ABSTRACT

Extracellular vesicles (EVs) are cell-derived membrane-bound structures that are believed to play a major role in intercellular communication by allowing cells to exchange proteins and genetic cargo between them. In particular, pathogens, such as the malaria parasite Plasmodium (P.) falciparum, utilize EVs to promote their growth and to alter their host's response. Thus, better characterization of these secreted organelles will enhance our understanding of the cellular processes that govern EVs' biology and pathological functions. Here we present a method that utilizes a high-end flow cytometer system to characterize small EVs, i.e., with a diameter less than 200 nm. Using this method, we could evaluate different parasite-derived EV populations according to their distinct cargo by using antibody-free labeling. It further allows to closely monitor a sub-population of vesicles carrying parasitic DNA cargo. This ability paves the way to conducting a more 'educated' analysis of the various EV cargo components.

10.
Cell Microbiol ; 21(7): e13021, 2019 07.
Article in English | MEDLINE | ID: mdl-30835870

ABSTRACT

Protozoan pathogens secrete nanosized particles called extracellular vesicles (EVs) to facilitate their survival and chronic infection. Here, we show the inhibition by Plasmodium berghei NK65 blood stage-derived EVs of the proliferative response of CD4+ T cells in response to antigen presentation. Importantly, these results were confirmed in vivo by the capacity of EVs to diminish the ovalbumin-specific delayed type hypersensitivity response. We identified two proteins associated with EVs, the histamine releasing factor (HRF) and the elongation factor 1α (EF-1α) that were found to have immunosuppressive activities. Interestingly, in contrast to WT parasites, EVs from genetically HRF- and EF-1α-deficient parasites failed to inhibit T cell responses in vitro and in vivo. At the level of T cells, we demonstrated that EVs from WT parasites dephosphorylate key molecules (PLCγ1, Akt, and ERK) of the T cell receptor signalling cascade. Remarkably, immunisation with EF-1α alone or in combination with HRF conferred a long-lasting antiparasite protection and immune memory. In conclusion, we identified a new mechanism by which P. berghei-derived EVs exert their immunosuppressive functions by altering T cell responses. The identification of two highly conserved immune suppressive factors offers new conceptual strategies to overcome EV-mediated immune suppression in malaria-infected individuals.


Subject(s)
Biomarkers, Tumor/genetics , Extracellular Vesicles/immunology , Malaria/genetics , Peptide Elongation Factor 1/genetics , Animals , Antigen Presentation/immunology , Antigens/genetics , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , Cell Proliferation/genetics , Extracellular Vesicles/genetics , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Malaria/parasitology , Malaria/pathology , Plasmodium berghei/genetics , Plasmodium berghei/pathogenicity , T-Lymphocytes/immunology , T-Lymphocytes/parasitology , Tumor Protein, Translationally-Controlled 1
12.
Front Immunol ; 9: 1011, 2018.
Article in English | MEDLINE | ID: mdl-29881375

ABSTRACT

Extracellular vesicles are essential for long distance cell-cell communication. They function as carriers of different compounds, including proteins, lipids and nucleic acids. Pathogens, like malaria parasites (Plasmodium falciparum, Pf), excel in employing vesicle release to mediate cell communication in diverse processes, particularly in manipulating the host response. Establishing research tools to study the interface between pathogen-derived vesicles and their host recipient cells will greatly benefit the scientific community. Here, we present an imaging flow cytometry (IFC) method for monitoring the uptake of malaria-derived vesicles by host immune cells. By staining different cargo components, we were able to directly track the cargo's internalization over time and measure the kinetics of its delivery. Impressively, we demonstrate that this method can be used to specifically monitor the translocation of a specific protein within the cellular milieu upon internalization of parasitic cargo; namely, we were able to visually observe how uptaken parasitic Pf-DNA cargo leads to translocation of transcription factor IRF3 from the cytosol to the nucleus within the recipient immune cell. Our findings demonstrate that our method can be used to study cellular dynamics upon vesicle uptake in different host-pathogen and pathogen-pathogen systems.


Subject(s)
Extracellular Vesicles/metabolism , Flow Cytometry , Interferon Regulatory Factor-3/metabolism , Monocytes/metabolism , Plasmodium falciparum , Biological Transport , DNA, Protozoan/metabolism , Host-Pathogen Interactions , Humans , Kinetics , Molecular Imaging , THP-1 Cells
13.
Nat Commun ; 8(1): 1985, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29215015

ABSTRACT

STING is an innate immune cytosolic adaptor for DNA sensors that engage malaria parasite (Plasmodium falciparum) or other pathogen DNA. As P. falciparum infects red blood cells and not leukocytes, how parasite DNA reaches such host cytosolic DNA sensors in immune cells is unclear. Here we show that malaria parasites inside red blood cells can engage host cytosolic innate immune cell receptors from a distance by secreting extracellular vesicles (EV) containing parasitic small RNA and genomic DNA. Upon internalization of DNA-harboring EVs by human monocytes, P. falciparum DNA is released within the host cell cytosol, leading to STING-dependent DNA sensing. STING subsequently activates the kinase TBK1, which phosphorylates the transcription factor IRF3, causing IRF3 to translocate to the nucleus and induce STING-dependent gene expression. This DNA-sensing pathway may be an important decoy mechanism to promote P. falciparum virulence and thereby may affect future strategies to treat malaria.


Subject(s)
Cytosol/immunology , DNA, Protozoan/immunology , Extracellular Vesicles/immunology , Malaria, Falciparum/immunology , Membrane Proteins/immunology , Plasmodium falciparum/immunology , Cell Line , Cell Nucleus/metabolism , Cryoelectron Microscopy , Cytosol/metabolism , DNA, Protozoan/metabolism , Erythrocytes , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/ultrastructure , Humans , Immunity, Innate , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Malaria, Falciparum/parasitology , Membrane Proteins/metabolism , Monocytes , Phosphorylation , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Primary Cell Culture , Protein Serine-Threonine Kinases/metabolism , RNA, Protozoan/immunology , RNA, Protozoan/metabolism , Signal Transduction
14.
Aging Cell ; 16(4): 661-671, 2017 08.
Article in English | MEDLINE | ID: mdl-28455874

ABSTRACT

Senescent cells are present in premalignant lesions and sites of tissue damage and accumulate in tissues with age. In vivo identification, quantification and characterization of senescent cells are challenging tasks that limit our understanding of the role of senescent cells in diseases and aging. Here, we present a new way to precisely quantify and identify senescent cells in tissues on a single-cell basis. The method combines a senescence-associated beta-galactosidase assay with staining of molecular markers for cellular senescence and of cellular identity. By utilizing technology that combines flow cytometry with high-content image analysis, we were able to quantify senescent cells in tumors, fibrotic tissues, and tissues of aged mice. Our approach also yielded the finding that senescent cells in tissues of aged mice are larger than nonsenescent cells. Thus, this method provides a basis for quantitative assessment of senescent cells and it offers proof of principle for combination of different markers of senescence. It paves the way for screening of senescent cells for identification of new senescence biomarkers, genes that bypass senescence or senolytic compounds that eliminate senescent cells, thus enabling a deeper understanding of the senescent state in vivo.


Subject(s)
Aging/genetics , Cellular Senescence/genetics , Neoplasms/genetics , Single-Cell Analysis/methods , Staining and Labeling/methods , Aging/metabolism , Aging/pathology , Animals , Biomarkers/analysis , Cellular Senescence/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Etoposide/pharmacology , Fibrosis , Flow Cytometry , Gene Expression , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Histones/genetics , Histones/metabolism , Humans , Image Processing, Computer-Assisted , Lymphocytes/metabolism , Lymphocytes/pathology , Mice , Molecular Imaging , Neoplasms/metabolism , Neoplasms/pathology , Primary Cell Culture , Stromal Cells/metabolism , Stromal Cells/pathology , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...