Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Immunity ; 47(3): 498-509.e6, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28916264

ABSTRACT

Double-stranded RNA (dsRNA) is a common by-product of viral infections and acts as a potent trigger of antiviral immunity. In the nematode C. elegans, sid-1 encodes a dsRNA transporter that is highly conserved throughout animal evolution, but the physiological role of SID-1 and its orthologs remains unclear. Here, we show that the mammalian SID-1 ortholog, SIDT2, is required to transport internalized extracellular dsRNA from endocytic compartments into the cytoplasm for immune activation. Sidt2-deficient mice exposed to extracellular dsRNA, encephalomyocarditis virus (EMCV), and herpes simplex virus 1 (HSV-1) show impaired production of antiviral cytokines and-in the case of EMCV and HSV-1-reduced survival. Thus, SIDT2 has retained the dsRNA transport activity of its C. elegans ortholog, and this transport is important for antiviral immunity.


Subject(s)
Immunity, Innate , Membrane Proteins/metabolism , RNA Transport , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/immunology , Cell Line , Cytoplasm , DEAD Box Protein 58/metabolism , Disease Models, Animal , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/immunology , Endosomes/metabolism , Female , Gene Expression , Gene Knockout Techniques , Herpes Simplex/genetics , Herpes Simplex/immunology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Lysosomes/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Nucleotide Transport Proteins , Protein Binding , Protein Transport , RNA, Viral/genetics , RNA, Viral/metabolism , Signal Transduction , Toll-Like Receptor 3/metabolism
2.
EMBO J ; 36(16): 2404-2418, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28716804

ABSTRACT

Type 2 inflammation is a defining feature of infection with parasitic worms (helminths), as well as being responsible for widespread suffering in allergies. However, the precise mechanisms involved in T helper (Th) 2 polarization by dendritic cells (DCs) are currently unclear. We have identified a previously unrecognized role for type I IFN (IFN-I) in enabling this process. An IFN-I signature was evident in DCs responding to the helminth Schistosoma mansoni or the allergen house dust mite (HDM). Further, IFN-I signaling was required for optimal DC phenotypic activation in response to helminth antigen (Ag), and efficient migration to, and localization with, T cells in the draining lymph node (dLN). Importantly, DCs generated from Ifnar1-/- mice were incapable of initiating Th2 responses in vivo These data demonstrate for the first time that the influence of IFN-I is not limited to antiviral or bacterial settings but also has a central role to play in DC initiation of Th2 responses.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/metabolism , Th2 Cells/immunology , Allergens/immunology , Animals , Mice , Mice, Knockout , Pyroglyphidae/immunology , Receptor, Interferon alpha-beta/deficiency , Schistosoma mansoni/immunology
3.
Immunol Cell Biol ; 98(1): 42-53, 2020 01.
Article in English | MEDLINE | ID: mdl-31559654

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) presents an increasing threat to public health, with antimicrobial resistance on the rise and infections endemic in the hospital setting. Despite a global research effort to understand and combat antimicrobial resistance, less work has focused on understanding the nuances in the immunopathogenesis of clinical strains. In particular, there is a surprising gap of knowledge in the literature pertaining to how clinical strains are recognized by dendritic cells (DCs). Here, we show that the activation of DCs is compromised in response to MRSA strains resistant to the last-line antibiotic daptomycin. We found a significant reduction in the secretion of proinflammatory cytokines including tumor necrosis factor-α, interleukin-6, regulated upon activation, normal T cell expressed, and secreted and macrophage inflammatory protein-1ß, as well as decreased expression of CD80 by DCs responding to daptomycin-resistant MRSA. We further demonstrate that this phenotype is coincident with the acquisition of specific point mutations in the cardiolipin synthase gene cls2, and, partly, in the bifunctional lysylphosphatidylglycerol flippase/synthetase gene mprF, which are genes that are often mutated in clinical daptomycin-resistant strains. Therefore, throughout infection and antibiotic therapy, MRSA has the capacity to not only develop further antibiotic resistance, but also develop resistance to immunological recognition by DCs, because of single amino acid point mutations occurring under the selective pressures of both host immunity and antibiotic therapy. Understanding the diversity of clinical MRSA isolates and the nuances in their immune recognition will have important implications for future therapeutics and the treatment of these infections.


Subject(s)
Daptomycin , Dendritic Cells/immunology , Drug Resistance, Bacterial/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Animals , B7-1 Antigen/immunology , Cytokines/immunology , Gene Expression Regulation , Humans , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Mice
4.
EMBO J ; 33(6): 542-58, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24514026

ABSTRACT

The sensing of nucleic acids by receptors of the innate immune system is a key component of antimicrobial immunity. RNA:DNA hybrids, as essential intracellular replication intermediates generated during infection, could therefore represent a class of previously uncharacterised pathogen-associated molecular patterns sensed by pattern recognition receptors. Here we establish that RNA:DNA hybrids containing viral-derived sequences efficiently induce pro-inflammatory cytokine and antiviral type I interferon production in dendritic cells. We demonstrate that MyD88-dependent signalling is essential for this cytokine response and identify TLR9 as a specific sensor of RNA:DNA hybrids. Hybrids therefore represent a novel molecular pattern sensed by the innate immune system and so could play an important role in host response to viruses and the pathogenesis of autoimmune disease.


Subject(s)
Dendritic Cells/metabolism , Immunity, Innate/immunology , Models, Immunological , Nucleic Acid Heteroduplexes/metabolism , Signal Transduction/immunology , Toll-Like Receptor 9/metabolism , Animals , Blotting, Western , Dendritic Cells/immunology , Endosomes , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescence Polarization , Fluorescent Antibody Technique , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/immunology , Nucleic Acid Heteroduplexes/immunology , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 9/immunology
5.
Immunol Cell Biol ; 94(4): 400-10, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26657145

ABSTRACT

Dendritic cells (DCs) are the key initiators of T-helper (Th) 2 immune responses against the parasitic helminth Schistosoma mansoni. Although the liver is one of the main sites of antigen deposition during infection with this parasite, it is not yet clear how distinct DC subtypes in this tissue respond to S. mansoni antigens in vivo, or how the liver microenvironment might influence DC function during establishment of the Th2 response. In this study, we show that hepatic DC subsets undergo distinct activation processes in vivo following murine infection with S. mansoni. Conventional DCs (cDCs) from schistosome-infected mice upregulated expression of the costimulatory molecule CD40 and were capable of priming naive CD4(+) T cells, whereas plasmacytoid DCs (pDCs) upregulated expression of MHC class II, CD86 and CD40 but were unable to support the expansion of either naive or effector/memory CD4(+) T cells. Importantly, in vivo depletion of pDCs revealed that this subset was dispensable for either maintenance or regulation of the hepatic Th2 effector response during acute S. mansoni infection. Our data provides strong evidence that S. mansoni infection favors the establishment of an immunogenic, rather than tolerogenic, liver microenvironment that conditions cDCs to initiate and maintain Th2 immunity in the context of ongoing antigen exposure.


Subject(s)
Dendritic Cells/immunology , Liver/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Th2 Cells/immunology , Animals , Antigens, Helminth/immunology , Cell Differentiation , Cells, Cultured , Dendritic Cells/parasitology , Liver/parasitology , Lymphocyte Activation , Mice , Mice, Inbred C57BL
6.
Nature ; 459(7249): 945-9, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19536257

ABSTRACT

Several hundred malaria parasite proteins are exported beyond an encasing vacuole and into the cytosol of the host erythrocyte, a process that is central to the virulence and viability of the causative Plasmodium species. The trafficking machinery responsible for this export is unknown. Here we identify in Plasmodium falciparum a translocon of exported proteins (PTEX), which is located in the vacuole membrane. The PTEX complex is ATP-powered, and comprises heat shock protein 101 (HSP101; a ClpA/B-like ATPase from the AAA+ superfamily, of a type commonly associated with protein translocons), a novel protein termed PTEX150 and a known parasite protein, exported protein 2 (EXP2). EXP2 is the potential channel, as it is the membrane-associated component of the core PTEX complex. Two other proteins, a new protein PTEX88 and thioredoxin 2 (TRX2), were also identified as PTEX components. As a common portal for numerous crucial processes, this translocon offers a new avenue for therapeutic intervention.


Subject(s)
Malaria, Falciparum/parasitology , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Animals, Genetically Modified , Models, Biological , Protein Binding , Protein Transport
7.
J Immunol ; 188(5): 2350-8, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22287718

ABSTRACT

Infection with schistosome helminths is associated with granulomatous inflammation that forms around parasite eggs trapped in host tissues. In severe cases, the resulting fibrosis can lead to organ failure, portal hypertension, and fatal bleeding. Murine studies identified IL-17 as a critical mediator of this immunopathology, and mouse strains that produce high levels of IL-17 in response to schistosome infection show increased mortality. In this article, we demonstrate that schistosome-specific IL-17 induction by dendritic cells from low-pathology C57BL/6 mice is normally regulated by their concomitant induction of IL-10. Simultaneous stimulation of schistosome-exposed C57BL/6 dendritic cells with a heat-killed bacterium enabled these cells to overcome IL-10 regulation and induce IL-17, even in wild-type C57BL/6 recipients. This schistosome-specific IL-17 was dependent on IL-6 production by the copulsed dendritic cells. Coimmunization of C57BL/6 animals with bacterial and schistosome Ags also resulted in schistosome-specific IL-17, and this response was enhanced in the absence of IL-10-mediated immune regulation. Together, our data suggest that the balance of pro- and anti-inflammatory cytokines that determines the severity of pathology during schistosome infection can be influenced not only by host and parasite, but also by concurrent bacterial stimulation.


Subject(s)
Antigens, Helminth/physiology , Dendritic Cells/immunology , Interleukin-17/biosynthesis , Propionibacterium acnes/immunology , Adoptive Transfer , Animals , Antigens, Helminth/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Interleukin-10/metabolism , Interleukin-10/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Schistosoma mansoni/immunology , Schistosomiasis/immunology , Schistosomiasis/metabolism , Schistosomiasis/pathology
9.
J Immunol ; 185(6): 3632-42, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20720206

ABSTRACT

Cerebral malaria is a severe complication of malaria. Sequestration of parasitized RBCs in brain microvasculature is associated with disease pathogenesis, but our understanding of this process is incomplete. In this study, we examined parasite tissue sequestration in an experimental model of cerebral malaria (ECM). We show that a rapid increase in parasite biomass is strongly associated with the induction of ECM, mediated by IFN-gamma and lymphotoxin alpha, whereas TNF and IL-10 limit this process. Crucially, we discovered that host CD4(+) and CD8(+) T cells promote parasite accumulation in vital organs, including the brain. Modulation of CD4(+) T cell responses by helminth coinfection amplified CD4(+) T cell-mediated parasite sequestration, whereas vaccination could generate CD4(+) T cells that reduced parasite biomass and prevented ECM. These findings provide novel insights into immune-mediated mechanisms of ECM pathogenesis and highlight the potential of T cells to both prevent and promote infectious diseases.


Subject(s)
Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Plasmodium berghei/immunology , Animals , Brain/blood supply , Brain/immunology , Brain/parasitology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , CD4-Positive T-Lymphocytes/pathology , Disease Models, Animal , Erythrocytes/immunology , Erythrocytes/parasitology , Erythrocytes/pathology , Female , Gastrointestinal Tract/blood supply , Gastrointestinal Tract/immunology , Gastrointestinal Tract/parasitology , Kidney/blood supply , Kidney/immunology , Kidney/parasitology , Liver/blood supply , Liver/immunology , Liver/parasitology , Lung/blood supply , Lung/immunology , Lung/parasitology , Malaria, Cerebral/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Organ Specificity/immunology , Plasmodium berghei/growth & development , Severity of Illness Index , Spleen/blood supply , Spleen/immunology , Spleen/parasitology
10.
Front Immunol ; 13: 906338, 2022.
Article in English | MEDLINE | ID: mdl-35958580

ABSTRACT

Schistosomiasis is a disease of global significance, with severity and pathology directly related to how the host responds to infection. The immunological narrative of schistosomiasis has been constructed through decades of study, with researchers often focussing on isolated time points, cell types and tissue sites of interest. However, the field currently lacks a comprehensive and up-to-date understanding of the immune trajectory of schistosomiasis over infection and across multiple tissue sites. We have defined schistosome-elicited immune responses at several distinct stages of the parasite lifecycle, in three tissue sites affected by infection: the liver, spleen, and mesenteric lymph nodes. Additionally, by performing RNA-seq on the livers of schistosome infected mice, we have generated novel transcriptomic insight into the development of schistosome-associated liver pathology and fibrosis across the breadth of infection. Through depletion of CD11c+ cells during peak stages of schistosome-driven inflammation, we have revealed a critical role for CD11c+ cells in the co-ordination and regulation of Th2 inflammation during infection. Our data provide an updated and high-resolution account of how host immune responses evolve over the course of murine schistosomiasis, underscoring the significance of CD11c+ cells in dictating host immunopathology against this important helminth infection.


Subject(s)
Schistosomiasis mansoni , Schistosomiasis , Animals , CD11c Antigen , Immunity , Inflammation , Mice , Schistosoma mansoni
11.
Eur J Immunol ; 40(6): 1674-81, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20391433

ABSTRACT

Despite extensive evidence that Plasmodium species are capable of stimulating the immune system, the association of malaria with a higher incidence of other infectious diseases and reduced responses to vaccination against unrelated pathogens suggests the existence of immune suppression. Recently, we provided evidence that blood-stage Plasmodium berghei infection leads to suppression of MHC class I-restricted immunity to third party (non-malarial) antigens as a consequence of systemic DC activation. This earlier study did not, however, determine whether reactivity was also impaired to MHC class II-restricted third party antigens or to Plasmodium antigens themselves. Here, we show that while P. berghei-expressed antigens were presented early in infection, there was a rapid decline in presentation within 4 days, paralleling impairment in MHC class I- and II-restricted presentation of third party antigens. This provides important evidence that P. berghei not only causes immunosuppression to subsequently encountered third party antigens, but also rapidly limits the capacity to generate effective parasite-specific immunity.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Immune Tolerance/immunology , Malaria/immunology , Animals , Antigens, Protozoan/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Parasites/immunology , Plasmodium berghei/immunology
12.
PLoS Pathog ; 5(4): e1000369, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19343215

ABSTRACT

Plasmodium falciparum malaria causes 660 million clinical cases with over 2 million deaths each year. Acquired host immunity limits the clinical impact of malaria infection and provides protection against parasite replication. Experimental evidence indicates that cell-mediated immune responses also result in detrimental inflammation and contribute to severe disease induction. In both humans and mice, the spleen is a crucial organ involved in blood stage malaria clearance, while organ-specific disease appears to be associated with sequestration of parasitized erythrocytes in vascular beds and subsequent recruitment of inflammatory leukocytes. Using a rodent model of cerebral malaria, we have previously found that the majority of T lymphocytes in intravascular infiltrates of cerebral malaria-affected mice express the chemokine receptor CXCR3. Here we investigated the effect of IP-10 blockade in the development of experimental cerebral malaria and the induction of splenic anti-parasite immunity. We found that specific neutralization of IP-10 over the course of infection and genetic deletion of this chemokine in knockout mice reduces cerebral intravascular inflammation and is sufficient to protect P. berghei ANKA-infected mice from fatality. Furthermore, our results demonstrate that lack of IP-10 during infection significantly reduces peripheral parasitemia. The increased resistance to infection observed in the absence of IP-10-mediated cell trafficking was associated with retention and subsequent expansion of parasite-specific T cells in spleens of infected animals, which appears to be advantageous for the control of parasite burden. Thus, our results demonstrate that modulating homing of cellular immune responses to malaria is critical for reaching a balance between protective immunity and immunopathogenesis.


Subject(s)
Chemokine CXCL10/immunology , Malaria, Cerebral/immunology , Malaria/immunology , Plasmodium berghei/physiology , T-Lymphocytes/immunology , Animals , Brain Chemistry , Chemokine CXCL10/genetics , Disease Models, Animal , Immunity, Cellular , Inflammation , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutralization Tests , Parasitemia/metabolism , Spleen/immunology , Statistics, Nonparametric
13.
Blood ; 113(14): 3352-62, 2009 Apr 02.
Article in English | MEDLINE | ID: mdl-19179303

ABSTRACT

Insights into the role of ankyrin-1 (ANK-1) in the formation and stabilization of the red cell cytoskeleton have come from studies on the nb/nb mice, which carry hypomorphic alleles of Ank-1. Here, we revise several paradigms established in the nb/nb mice through analysis of an N-ethyl-N-nitrosourea (ENU)-induced Ank-1-null mouse. Mice homozygous for the Ank-1 mutation are profoundly anemic in utero and most die perinatally, indicating that Ank-1 plays a nonredundant role in erythroid development. The surviving pups exhibit features of severe hereditary spherocytosis (HS), with marked hemolysis, jaundice, compensatory extramedullary erythropoiesis, and tissue iron overload. Red cell membrane analysis reveals a complete loss of ANK-1 protein and a marked reduction in beta-spectrin. As a consequence, the red cells exhibit total disruption of cytoskeletal architecture and severely altered hemorheologic properties. Heterozygous mutant mice, which have wild-type levels of ANK-1 and spectrin in their RBC membranes and normal red cell survival and ultrastructure, exhibit profound resistance to malaria, which is not due to impaired parasite entry into RBC. These findings provide novel insights into the role of Ank-1, and define an ideal model for the study of HS and malarial resistance.


Subject(s)
Ankyrins/physiology , Erythroid Cells/metabolism , Ethylnitrosourea , Hematologic Neoplasms/chemically induced , Hematologic Neoplasms/genetics , Animals , Animals, Newborn , Ankyrins/genetics , Ankyrins/metabolism , Base Sequence , Carcinogens , Cytoskeleton/genetics , Cytoskeleton/pathology , DNA Mutational Analysis , Erythrocytes/drug effects , Erythrocytes/pathology , Erythrocytes, Abnormal/pathology , Erythropoiesis/genetics , Erythropoiesis/physiology , Hematologic Neoplasms/pathology , Hemolysis/drug effects , Hemolysis/genetics , Malaria/genetics , Malaria/veterinary , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data
14.
Proc Natl Acad Sci U S A ; 105(38): 14509-14, 2008 Sep 23.
Article in English | MEDLINE | ID: mdl-18799734

ABSTRACT

Although CD8(+) T cells do not contribute to protection against the blood stage of Plasmodium infection, there is mounting evidence that they are principal mediators of murine experimental cerebral malaria (ECM). At present, there is no direct evidence that the CD8(+) T cells mediating ECM are parasite-specific or, for that matter, whether parasite-specific CD8(+) T cells are generated in response to blood-stage infection. To resolve this and to define the cellular requirements for such priming, we generated transgenic P. berghei parasites expressing model T cell epitopes. This approach was necessary as MHC class I-restricted antigens to blood-stage infection have not been defined. Here, we show that blood-stage infection leads to parasite-specific CD8(+) and CD4(+) T cell responses. Furthermore, we show that P. berghei-expressed antigens are cross-presented by the CD8alpha(+) subset of dendritic cells (DC), and that this induces pathogen-specific cytotoxic T lymphocytes (CTL) capable of lysing cells presenting antigens expressed by blood-stage parasites. Finally, using three different experimental approaches, we provide evidence that CTL specific for parasite-expressed antigens contribute to ECM.


Subject(s)
Antigens, Protozoan/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Plasmodium berghei/immunology , Animals , Animals, Genetically Modified , Brain/immunology , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/immunology , Life Cycle Stages , Malaria, Cerebral/blood , Malaria, Cerebral/mortality , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Plasmodium berghei/genetics , Plasmodium berghei/growth & development
15.
Immunohorizons ; 5(8): 721-732, 2021 08 30.
Article in English | MEDLINE | ID: mdl-34462311

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are potent producers of type I IFN (IFN-I) during viral infection and respond to IFN-I in a positive feedback loop that promotes their function. IFN-I shapes dendritic cell responses during helminth infection, impacting their ability to support Th2 responses. However, the role of pDCs in type 2 inflammation is unclear. Previous studies have shown that pDCs are dispensable for hepatic or splenic Th2 responses during the early stages of murine infection with the trematode Schistosoma mansoni at the onset of parasite egg laying. However, during S. mansoni infection, an ongoing Th2 response against mature parasite eggs is required to protect the liver and intestine from acute damage and how pDCs participate in immune responses to eggs and adult worms in various tissues beyond acute infection remains unclear. We now show that pDCs are required for optimal Th2 cytokine production in response to S. mansoni eggs in the intestinal-draining mesenteric lymph nodes throughout infection and for egg-specific IFN-γ at later time points of infection. Further, pDC depletion at chronic stages of infection led to increased hepatic and splenic pathology as well as abrogated Th2 cell cytokine production and activation in the liver. In vitro, mesenteric lymph node pDCs supported Th2 cell responses from infection-experienced CD4+ T cells, a process dependent on pDC IFN-I responsiveness, yet independent of Ag. Together, these data highlight a previously unappreciated role for pDCs and IFN-I in maintaining and reinforcing type 2 immunity in the lymph nodes and inflamed tissue during helminth infection.


Subject(s)
Cytokines/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/parasitology , Cytokines/metabolism , Dendritic Cells/parasitology , Female , Flow Cytometry/methods , Host-Parasite Interactions/immunology , Lymphocyte Count , Mice, Inbred C57BL , Mice, Knockout , Schistosoma mansoni/physiology , Schistosomiasis mansoni/metabolism , Schistosomiasis mansoni/parasitology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/parasitology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/parasitology
16.
Front Microbiol ; 11: 702, 2020.
Article in English | MEDLINE | ID: mdl-32373101

ABSTRACT

Severe malaria anemia is one of the most common causes of morbidity and mortality arising from infection with Plasmodium falciparum. The pathogenesis of malarial anemia is complex, involving both parasite and host factors. As mouse models of malaria also develop anemia, they can provide a useful resource to study the impact of Plasmodium infections and the resulting host innate immune response on erythropoiesis. In this study, we have characterized the bone marrow and splenic responses of the erythroid as well as other hematopoietic lineages after an acute infection of Balb/c mice with Plasmodium berghei. Such characterization of the hematopoietic changes is critical to underpin future studies, using knockout mice and transgenic parasites, to tease out the interplay between host genes and parasite modulators implicated in susceptibility to malaria anemia. P. berghei infection led to a clear perturbation of steady-state erythropoiesis, with the most profound defects in polychromatic and orthochromatic erythroblasts as well as erythroid colony- and burst-forming units (CFU-E and BFU-E), resulting in an inability to compensate for anemia. The perturbation in erythropoiesis was not attributable to parasites infecting erythroblasts and affecting differentiation, nor to insufficient erythropoietin (EPO) production or impaired activation of the Signal transducer and activator of transcription 5 (STAT5) downstream of the EPO receptor, indicating EPO-signaling remained functional in anemia. Instead, the results point to acute anemia in P. berghei-infected mice arising from increased myeloid cell production in order to clear the infection, and the concomitant release of pro-inflammatory cytokines and chemokines from myeloid cells that inhibit erythroid development, in a manner that resembles the pathophysiology of anemia of chronic disease.

17.
Clin Transl Immunology ; 8(11): e01083, 2019.
Article in English | MEDLINE | ID: mdl-31700626

ABSTRACT

This article summarises recent advances reported at the 9th Lorne Infection and Immunity Conference. This exciting conference hosted speakers in the fields of innate and adaptive responses to infection including host-pathogen interactions as well as novel strategies for the detection, control and treatment of infectious diseases such as fluorescent antibiotics and vaccine development. Host-pathogen studies focused on a broad range of pathogens including malaria, CMV, influenza, dengue and Zika viruses, listeria and tuberculosis.

18.
Front Immunol ; 10: 357, 2019.
Article in English | MEDLINE | ID: mdl-30886619

ABSTRACT

Malaria remains a serious threat to global health. Sustained malaria control and, eventually, eradication will only be achieved with a broadly effective malaria vaccine. Yet a fundamental lack of knowledge about how antimalarial immunity is acquired has hindered vaccine development efforts to date. Understanding how malaria-causing parasites modulate the host immune system, specifically dendritic cells (DCs), key initiators of adaptive and vaccine antigen-based immune responses, is vital for effective vaccine design. This review comprehensively summarizes how exposure to Plasmodium spp. impacts human DC function in vivo and in vitro. We have highlighted the heterogeneity of the data observed in these studies, compared and critiqued the models used to generate our current understanding of DC function in malaria, and examined the mechanisms by which Plasmodium spp. mediate these effects. This review highlights potential research directions which could lead to improved efficacy of existing vaccines, and outlines novel targets for next-generation vaccine strategies to target malaria.


Subject(s)
Antigens, Protozoan/immunology , Dendritic Cells/immunology , Malaria Vaccines/immunology , Malaria/immunology , Plasmodium/immunology , Humans , Malaria/prevention & control , Malaria Vaccines/therapeutic use
19.
Front Immunol ; 10: 32, 2019.
Article in English | MEDLINE | ID: mdl-30766530

ABSTRACT

Dendritic cells are key linkers of innate and adaptive immunity. Efficient dendritic cell activation is central to the acquisition of immunity and the efficacy of vaccines. Understanding how dendritic cells are affected by Plasmodium falciparum blood-stage parasites will help to understand how immunity is acquired and maintained, and how vaccine responses may be impacted by malaria infection or exposure. This study investigates the response of dendritic cells to two different life stages of the malaria parasite, parasitized red blood cells and merozoites, using a murine model. We demonstrate that the dendritic cell responses to merozoites are robust whereas dendritic cell activation, particularly CD40 and pro-inflammatory cytokine expression, is compromised in the presence of freshly isolated parasitized red blood cells. The mechanism of dendritic cell suppression by parasitized red blood cells is host red cell membrane-independent. Furthermore, we show that cryopreserved parasitized red blood cells have a substantially reduced capacity for dendritic cell activation.


Subject(s)
Dendritic Cells/immunology , Host-Parasite Interactions/immunology , Life Cycle Stages/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium falciparum/immunology , Biomarkers , Cytokines/metabolism , Dendritic Cells/metabolism , Erythrocytes/immunology , Erythrocytes/parasitology , Humans , Ligands , Plasmodium falciparum/growth & development , Toll-Like Receptor 9/metabolism
20.
Nat Commun ; 6: 6920, 2015 Apr 24.
Article in English | MEDLINE | ID: mdl-25908537

ABSTRACT

Dendritic cells (DCs) direct CD4(+) T-cell differentiation into diverse helper (Th) subsets that are required for protection against varied infections. However, the mechanisms used by DCs to promote Th2 responses, which are important both for immunity to helminth infection and in allergic disease, are currently poorly understood. We demonstrate a key role for the protein methyl-CpG-binding domain-2 (Mbd2), which links DNA methylation to repressive chromatin structure, in regulating expression of a range of genes that are associated with optimal DC activation and function. In the absence of Mbd2, DCs display reduced phenotypic activation and a markedly impaired capacity to initiate Th2 immunity against helminths or allergens. These data identify an epigenetic mechanism that is central to the activation of CD4(+) T-cell responses by DCs, particularly in Th2 settings, and reveal methyl-CpG-binding proteins and the genes under their control as possible therapeutic targets for type-2 inflammation.


Subject(s)
DNA-Binding Proteins/immunology , Dendritic Cells/immunology , Gene Expression Regulation/genetics , RNA, Messenger/metabolism , Th2 Cells/immunology , Allergens , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Polarity , Chromatin Immunoprecipitation , DNA Methylation , DNA-Binding Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Epigenesis, Genetic , Flow Cytometry , Hypersensitivity/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Pyroglyphidae/immunology , Reverse Transcriptase Polymerase Chain Reaction , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology
SELECTION OF CITATIONS
SEARCH DETAIL