Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 874308, 2022.
Article in English | MEDLINE | ID: mdl-35757733

ABSTRACT

Infection with helminths or parasitic worms are highly prevalent worldwide especially in developing regions. Helminths cause chronic infections that are associated with suppression of immune responses to unrelated pathogens, vaccines, and by-stander antigens responsible for dysregulated immune responses as occurs in diseases such as allergies. Helminths use multiple mechanisms to modulate the immune system to evade the highly polarized type 2 immune response required to expel adult worms and for immunity to reinfection. Anthelmintic drugs are efficient in reducing adult worm burdens in helminth-infected individuals, but resistance to these drugs is rapidly increasing and vaccines against these pathogens are not available. Emerging evidence indicate that helminths induce myeloid-derived suppressor cells (MDSC), originally described in tumor-bearing mice and cancer patients. MDSC are a heterogenous population of immature cells that consist of two distinct sub-populations, polymorphonuclear (PMN)-MDSC and monocytic (M)-MDSC based on morphology and phenotype. MDSC suppress the function of T cells and other innate and adaptive immune cells including NK cells and B cells. During cancer or infection with bacteria or viruses, there is marked expansion of MDSC. Furthermore, the frequencies of MDSC correlate inversely with the prognosis and survival of tumor-bearing hosts as well as bacterial and viral burdens, persistence, and outcome in infected hosts. Currently, there is a paucity of data on MDSC and helminth infections. Here, we provide a survey of the evidence accumulated so far that overall support a role for MDSC in modulating immune responses during helminth infections. We review data from studies in various helminths, including those that infect humans. Finally, we summarize the progress to date in understanding the role of MDSC in helminth infections and briefly discuss potential host-directed strategies to target MDSC-mediated suppression of immune responses to helminths in favor of development of immunity to eliminate adult worms and possibly induce protection against reinfection.


Subject(s)
Helminths , Myeloid-Derived Suppressor Cells , Neoplasms , Vaccines , Animals , Humans , Mice , Reinfection , T-Lymphocytes
2.
Cell Rep ; 32(12): 108170, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32966787

ABSTRACT

The replication cycle and pathogenesis of the Plasmodium malarial parasite involves rapid expansion in red blood cells (RBCs), and variants of certain RBC-specific proteins protect against malaria in humans. In RBCs, bisphosphoglycerate mutase (BPGM) acts as a key allosteric regulator of hemoglobin/oxyhemoglobin. We demonstrate here that a loss-of-function mutation in the murine Bpgm (BpgmL166P) gene confers protection against both Plasmodium-induced cerebral malaria and blood-stage malaria. The malaria protection seen in BpgmL166P mutant mice is associated with reduced blood parasitemia levels, milder clinical symptoms, and increased survival. The protective effect of BpgmL166P involves a dual mechanism that enhances the host's stress erythroid response to Plasmodium-driven RBC loss and simultaneously alters the intracellular milieu of the RBCs, including increased oxyhemoglobin and reduced energy metabolism, reducing Plasmodium maturation, and replication. Overall, our study highlights the importance of BPGM as a regulator of hemoglobin/oxyhemoglobin in malaria pathogenesis and suggests a new potential malaria therapeutic target.


Subject(s)
Anemia/etiology , Anemia/prevention & control , Bisphosphoglycerate Mutase/deficiency , Malaria, Cerebral/enzymology , Malaria, Cerebral/prevention & control , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Bisphosphoglycerate Mutase/chemistry , Bisphosphoglycerate Mutase/genetics , Bisphosphoglycerate Mutase/metabolism , Enzyme Stability , Erythrocytes/enzymology , Erythrocytes/parasitology , Erythropoiesis , Extracellular Matrix/metabolism , Female , HEK293 Cells , Humans , Malaria, Cerebral/complications , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation/genetics , Parasites/growth & development , Plasmodium/growth & development , Polycythemia
3.
J Inorg Biochem ; 200: 110808, 2019 11.
Article in English | MEDLINE | ID: mdl-31487576

ABSTRACT

In malaria, Plasmodium parasites produce hemozoin (Hz) as a route to detoxify free heme released from the catabolism of hemoglobin. Hz isolated from the parasites is encapsulated in an organic layer constituted by parasite and host components. This organic coating may play a role in Hz formation and in the immunomodulatory properties attributed to Hz, and they may influence the mode of action of antimalarials that block Hz formation. In this work, we analyze the organic layer adhered to Hz, and find Na, Cl, Si, Ca and P present, in addition to organic material. Our results suggest that Na, Cl, and P adsorb during Hz release from the red blood cells, while Si and Ca derive from components present during Hz biomineralization within the digestive vacuole of the parasite. Overall, we show that inorganic elements associated with Hz surface provide insights into the biological functions of Plasmodium parasites.


Subject(s)
Erythrocytes , Hemeproteins/metabolism , Plasmodium chabaudi/metabolism , Plasmodium falciparum/metabolism , Animals , Erythrocytes/metabolism , Erythrocytes/parasitology , Humans , Ions/pharmacology , Mice
4.
Sci Rep ; 8(1): 15921, 2018 10 29.
Article in English | MEDLINE | ID: mdl-30374177

ABSTRACT

Parasitic worms have a remarkable ability to modulate host immune responses through several mechanisms including excreted/secreted proteins (ESP), yet the exact nature of these proteins and their targets often remains elusive. Here, we performed mass spectrometry analyses of ESP (TsESP) from larval and adult stages of the pig whipworm Trichuris suis (Ts) and identified ~350 proteins. Transcriptomic analyses revealed large subsets of differentially expressed genes in the various life cycle stages of the parasite. Exposure of bone marrow-derived macrophages and dendritic cells to TsESP markedly diminished secretion of the pro-inflammatory cytokines TNFα and IL-12p70. Conversely, TsESP exposure strongly induced release of the anti-inflammatory cytokine IL-10, and also induced high levels of nitric oxide (NO) and upregulated arginase activity in macrophages. Interestingly, TsESP failed to directly induce CD4+ CD25+ FoxP3+ regulatory T cells (Treg cells), while OVA-pulsed TsESP-treated dendritic cells suppressed antigen-specific OT-II CD4+ T cell proliferation. Fractionation of TsESP identified a subset of proteins that promoted anti-inflammatory functions, an activity that was recapitulated using recombinant T. suis triosephosphate isomerase (TPI) and nucleoside diphosphate kinase (NDK). Our study helps illuminate the intricate balance that is characteristic of parasite-host interactions at the immunological interface, and further establishes the principle that specific parasite-derived proteins can modulate immune cell functions.


Subject(s)
Helminth Proteins/metabolism , Trichuris/metabolism , Animals , Arginase/metabolism , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Life Cycle Stages , Macrophages/cytology , Macrophages/metabolism , Nitric Oxide/metabolism , Swine/parasitology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Trichuris/growth & development
5.
PLoS Pathog ; 13(10): e1006647, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28968468

ABSTRACT

Interferon regulatory factor-8 (IRF-8) is critical for Th1 cell differentiation and negatively regulates myeloid cell development including myeloid-derived suppressor cells (MDSC). MDSC expand during infection with various pathogens including the gastrointestinal (GI) nematode Heligmosomoides polygyrus bakeri (Hpb). We investigated if IRF-8 contributes to Th2 immunity to Hpb infection. Irf8 expression was down-regulated in MDSC from Hpb-infected C57BL/6 (B6) mice. IRF-8 deficient Irf8-/- and BXH-2 mice had significantly higher adult worm burdens than B6 mice after primary or challenge Hpb infection. During primary infection, MDSC expanded to a significantly greater extent in mesenteric lymph nodes (MLN) and spleens of Irf8-/- and BXH-2 than B6 mice. CD4+GATA3+ T cells numbers were comparable in MLN of infected B6 and IRF-8 deficient mice, but MLN cells from infected IRF-8 deficient mice secreted significantly less parasite-specific IL-4 ex vivo. The numbers of alternatively activated macrophages in MLN and serum levels of Hpb-specific IgG1 and IgE were also significantly less in infected Irf8-/- than B6 mice. The frequencies of antigen-experienced CD4+CD11ahiCD49dhi cells that were CD44hiCD62L- were similar in MLN of infected Irf8-/- and B6 mice, but the proportions of CD4+GATA3+ and CD4+IL-4+ T cells were lower in infected Irf8-/- mice. CD11b+Gr1+ cells from naïve or infected Irf8-/- mice suppressed CD4+ T cell proliferation and parasite-specific IL-4 secretion in vitro albeit less efficiently than B6 mice. Surprisingly, there were significantly more CD4+ T cells in infected Irf8-/- mice, with a higher frequency of CD4+CD25+Foxp3+ T (Tregs) cells and significantly higher numbers of Tregs than B6 mice. In vivo depletion of MDSC and/or Tregs in Irf8-/- mice did not affect adult worm burdens, but Treg depletion resulted in higher egg production and enhanced parasite-specific IL-5, IL-13, and IL-6 secretion ex vivo. Our data thus provide a previously unrecognized role for IRF-8 in Th2 immunity to a GI nematode.


Subject(s)
Gastrointestinal Diseases/immunology , Interferon Regulatory Factors/immunology , Myeloid-Derived Suppressor Cells/immunology , Nematode Infections/immunology , Nematospiroides dubius/immunology , Th2 Cells/immunology , Animals , Cells, Cultured , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Interferon Regulatory Factors/drug effects , Interferon Regulatory Factors/genetics , Interleukin-4/metabolism , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/immunology
6.
Exp Parasitol ; 178: 30-36, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28533110

ABSTRACT

To better control gastrointestinal nematode infections in humans and animals, it is important to understand the strategies used by these parasites to modulate the host immune system. In this regard, molecules released by parasites have been attributed crucially important roles in host-parasite negotiations. We characterized the excretory/secretory (E/S) microRNA (miRNA) and protein profiles from the mouse gastrointestinal nematode parasite Trichuris muris. Released miRNAs were subjected to miRNA sequencing and E/S proteins were analysed by mass spectrometry. Fourteen miRNAs were identified in T. muris exosome-like vesicles, as well as 73 proteins of nematode origin, 11 of which were unique to this study. Comparison with published nematode protein secretomes revealed high conservation at the functional level.


Subject(s)
Exosomes/chemistry , Helminth Proteins/analysis , MicroRNAs/isolation & purification , Trichuris/metabolism , Animals , Culture Media/chemistry , Helminth Proteins/genetics , Helminth Proteins/immunology , Helminth Proteins/physiology , Proteomics , Reproducibility of Results , Trichuris/genetics , Trichuris/immunology
7.
PLoS One ; 12(5): e0177818, 2017.
Article in English | MEDLINE | ID: mdl-28542307

ABSTRACT

Pyruvate kinase (PKLR) deficiency protects mice and humans against blood-stage malaria. Although mouse strain AcB62 carries a malaria-protective PklrI90N genetic mutation, it is phenotypically susceptible to blood stage malaria induced by infection with Plasmodium chabaudi AS, suggesting a genetic modifier of the PklrI90N protective effect. Linkage analysis in a F2 cross between AcB62 (PklrI90N) and another PK deficient strain CBA/Pk (PklrG338D) maps this modifier (designated Char10) to chromosome 9 (LOD = 10.8, 95% Bayesian CI = 50.7-75Mb). To study the mechanistic basis of the Char10 effect, we generated an incipient congenic line (Char10C) that harbors the Char10 chromosome 9 segment from AcB62 fixed on the genetic background of CBA/Pk. The Char10 effect is shown to be highly penetrant as the Char10C line recapitulates the AcB62 phenotype, displaying high parasitemia following P. chabaudi infection, compared to CBA/Pk. Char10C mice also display a reduction in anemia phenotypes associated with the PklrG338D mutation including decreased splenomegaly, decreased circulating reticulocytes, increased density of mature erythrocytes, increased hematocrit, as well as decreased iron overload in kidney and liver and decreased serum iron. Erythroid lineage analyses indicate that the number of total TER119+ cells as well as the numbers of the different CD71+/CD44+ erythroblast sub-populations were all found to be lower in Char10C spleen compared to CBA/Pk. Char10C mice also displayed lower number of CFU-E per spleen compared to CBA/Pk. Taken together, these results indicate that the Char10 locus modulates the severity of pyruvate kinase deficiency by regulating erythroid responses in the presence of PK-deficiency associated haemolytic anemia.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Chromosomes, Mammalian/genetics , Genetic Loci/genetics , Genetic Predisposition to Disease/genetics , Malaria/genetics , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/metabolism , Anemia, Hemolytic, Congenital Nonspherocytic/physiopathology , Animals , Erythrocytes/metabolism , Erythrocytes/pathology , Erythropoiesis/genetics , Humans , Iron/metabolism , Mice , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Pyruvate Metabolism, Inborn Errors/metabolism , Pyruvate Metabolism, Inborn Errors/physiopathology
8.
Acta Biomater ; 48: 247-257, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27769943

ABSTRACT

Mucoadhesive drug delivery systems stick to mucosal tissues and prolong the local retention time of drugs. Since the colon is covered by a mucosal layer, mucoadhesive rectal formulations may improve treatment of such diseases as hypertension or colon cancer. Ulcerative colitis (UC) is an inflammatory bowel disease characterized by chronic inflammation of the colonic mucosa. It is commonly treated with sulfasalazine (SSZ), which is metabolized by the intestinal flora into the therapeutic 5-aminosalicylic acid (5-ASA) and a toxic by-product sulfapyridine (SP). SSZ can be administered orally or rectally. The latter route avoids unintended absorption of the drug or its degradation products in the upper gastrointestinal tract, but often fails due to limited retention time. Here, we propose a mucoadhesive hydrogel to improve the efficacy of rectal SSZ administration. The gel is made of catechol modified-chitosan (Cat-CS) crosslinked by genipin. After loading the gel with SSZ, we evaluated its efficacy in a mouse model of UC. Compared to oral SSZ treatment, rectal SSZ/Cat-CS delivery was more therapeutic, showed equivalent histological scores, and induced a lower plasma concentration of the potentially toxic SP by-product. These results show SSZ/Cat-CS rectal hydrogels are more effective and safer formulations for UC treatment than oral SSZ. STATEMENT OF SIGNIFICANCE: Ulcerative colitis affects the colon by causing chronic inflammation on the mucosa. One of the most common drugs to treat mild to moderate UC is sulfasalazine, which can be administrated both orally and rectally. Rectal formulations are preferable, since their therapeutic effect happens topically, and they prevent side effects related to absorption of the drug in the small intestine. However, the efficacy of rectal sulfasalazine formulations is decreased by their limited colon residence time. Here we propose a chitosan-catechol mucoadhesive gel that allows delivering sulfasalazine more effectively and safely than oral administration. Our results bring new insights into the field of mussel-inspired catechol hydrogels, showing their potential as drug delivery systems to treat a widespread disease such as ulcerative colitis.


Subject(s)
Adhesives/chemistry , Chitosan/chemistry , Colitis, Ulcerative/drug therapy , Drug Delivery Systems , Hydrogels/chemistry , Mucins/chemistry , Rectum/drug effects , Animals , Body Weight/drug effects , Catechols/chemistry , Colitis, Ulcerative/blood , Colitis, Ulcerative/pathology , Colon/drug effects , Colon/pathology , Dose-Response Relationship, Drug , Mice , Occult Blood , Sulfasalazine/blood , Sulfasalazine/metabolism , Sulfasalazine/pharmacology , Sulfasalazine/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
9.
Immunity ; 45(4): 831-846, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27760339

ABSTRACT

T follicular helper (Tfh) cells are a CD4+ T cell subset critical for long-lived humoral immunity. We hypothesized that integrins play a decisive role in Tfh cell biology. Here we show that Tfh cells expressed a highly active form of leukocyte function-associated antigen-1 (LFA-1) that was required for their survival within the germinal center niche. In addition, LFA-1 promoted expression of Bcl-6, a transcriptional repressor critical for Tfh cell differentiation, and inhibition of LFA-1 abolished Tfh cell generation and prevented protective humoral immunity to intestinal helminth infection. Furthermore, we demonstrated that expression of Talin-1, an adaptor protein that regulates LFA-1 affinity, dictated Tfh versus Th2 effector cell differentiation. Collectively, our results define unique functions for LFA-1 in the Tfh cell effector program and suggest that integrin activity is important in lineage decision-making events in the adaptive immune system.


Subject(s)
Cell Differentiation/immunology , Lymphocyte Activation/immunology , Lymphocyte Function-Associated Antigen-1/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cells, Cultured , Germinal Center/immunology , Humans , Immunity, Humoral/immunology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-6/immunology
10.
Malar J ; 15(1): 260, 2016 05 06.
Article in English | MEDLINE | ID: mdl-27150250

ABSTRACT

BACKGROUND: The potential emergence and spread of resistance to artemisinins in the Plasmodium falciparum malaria parasite constitutes a major global health threat. Hence, improving the efficacy of artemisinins and of artemisinin-based combination therapy (ACT) represents a major short-term goal in the global fight against malaria. Mice defective in the enzyme pantetheinase (Vnn3) show increased susceptibility to blood-stage malaria (increased parasitaemia, reduced survival), and supplementation of Vnn3 mutants with the reaction product of pantetheinase, cysteamine, corrects in part the malaria-susceptibility phenotype of the mutants. Cysteamine (Cys) is a small, naturally occurring amino-thiol that has very low toxicity in vivo and is approved for clinical use in the life-long treatment of the kidney disorder nephropathic cystinosis. METHODS: The ability of Cys to improve the anti-plasmodial activity of different clinically used artemisinins was tested. The effect of different CYS/ART combinations on malarial phenotypes (parasite blood-stage replication, overall and survival from lethal infection) was assessed in a series of in vivo experiments using Plasmodium strains that induce either blood-stage (Plasmodium chabaudi AS) or cerebral disease (Plasmodium berghei ANKA). This was also evaluated in an ex vivo experimental protocol that directly assesses the effect of such drug combinations on the viability of Plasmodium parasites, as measured by the ability of tested parasites to induce a productive infection in vivo in otherwise naïve animals. RESULTS: Cys is found to potentiate the anti-plasmodial activity of artesunate, artemether, and arteether, towards the blood-stage malaria parasite P. chabaudi AS. Ex vivo experiments, indicate that potentiation of the anti-plasmodial activity of artemisinins by Cys is direct and does not require the presence of host factors. In addition, potentiation occurs at sub-optimal concentrations of artemisinins and Cys that on their own have little or no effect on parasite growth. Cys also dramatically enhances the efficacy and protective effect of artemisinins against cerebral malaria induced by infection with the P. berghei ANKA parasite. CONCLUSION: These findings indicate that inclusion of Cys in current formulations of ACT, or its use as adjunct therapy could improve the anti-plasmodial activity of artemisinin, decrease mortality in cerebral malaria patients, and prevent or delay the development and spread of artemisinin resistance.


Subject(s)
Antimalarials/administration & dosage , Artemisinins/administration & dosage , Cysteamine/administration & dosage , Drug Synergism , Malaria/drug therapy , Animals , Cell Survival/drug effects , Disease Models, Animal , Drug Therapy, Combination , Female , Malaria, Cerebral/drug therapy , Mice , Plasmodium berghei/drug effects , Plasmodium berghei/physiology , Plasmodium chabaudi/drug effects , Plasmodium chabaudi/physiology , Survival Analysis , Treatment Outcome
11.
Nat Protoc ; 9(12): 2740-54, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25375989

ABSTRACT

Heligmosomoides polygyrus bakeri (Hpb) infection in mice is a convenient model for studying the pathophysiology and immunology of gastrointestinal (GI) helminth infection. Hpb infection suppresses immune responses to bystander antigens and unrelated pathogens, and it slows the progression and modifies the outcome of immune-mediated diseases. Hpb-derived excretory-secretory (ES) products potently modulate CD4(+) helper T cell (TH) responses by inducing regulatory T cells, tolerogenic dendritic cells (DCs) and immunoregulatory cytokines. This observation has spiked interest in identifying the immunomodulatory molecules, especially proteins, in ES products from Hpb and other GI nematodes for development as novel therapies to treat individuals with immune-mediated diseases, such as inflammatory bowel diseases (IBDs). In this protocol, we describe how to (i) maintain Hpb in the laboratory for experimental infections, (ii) collect adult worms from infected mice to generate ES products and (iii) evaluate the modulatory effects of ES products on toll-like receptor (TLR) ligand-induced maturation of CD11c(+) DCs. The three major sections of the PROCEDURE can be used independently, and they require ∼6, 10 and 27 h, respectively. Although other methods use a modified Baermann apparatus to collect Hpb adult worms, we describe a method that involves dissection of adult worms from intestinal tissue. The protocol will be useful to investigators studying the host-parasite interface and identifying and analyzing helminth-derived molecules with therapeutic potential.


Subject(s)
Immunologic Factors/pharmacology , Nematospiroides dubius/immunology , Nematospiroides dubius/isolation & purification , Strongylida Infections/immunology , Animals , CD11c Antigen/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Feces/parasitology , Female , Host-Parasite Interactions/immunology , Immunologic Factors/isolation & purification , Intestines/parasitology , Male , Mice , Mice, Inbred BALB C , Nematospiroides dubius/physiology , Strongylida Infections/parasitology
12.
J Infect Dis ; 209(1): 140-9, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23922378

ABSTRACT

Low reticulocytosis, indicating reduced red blood cell (RBC) output, is an important feature of severe malarial anemia. Evidence supports a role for Plasmodium products, especially hemozoin (Hz), in suppressed erythropoiesis during malaria, but the mechanism(s) involved remains unclear. Here, we demonstrated that low reticulocytosis and suppressed erythropoietin (Epo)-induced erythropoiesis are features of malarial anemia in Plasmodium yoelii- and Plasmodium berghei ANKA-infected mice, similar to our previous observations in Plasmodium chabaudi AS-infected mice. The magnitude of decreases in RBC was a reflection of parasitemia level, but low reticulocytosis was evident despite differences in parasitemia, clinical manifestation, and infection outcome. Schizont extracts and Hz from P. falciparum and P. yoelii and synthetic Hz suppressed Epo-induced proliferation of erythroid precursors in vitro but did not inhibit RBC maturation. To determine whether Hz contributes to malarial anemia, P. yoelii-derived or synthetic Hz was administered to naive mice, and the development of anemia, reticulocytosis, and RBC turnover was determined. Parasite-derived Hz induced significant decreases in RBC and increased RBC turnover with compensatory reticulocytosis, but anemia was not as severe as that in infected mice. Our findings suggest that parasite factors, including Hz, contribute to severe malarial anemia by suppressing Epo-induced proliferation of erythroid precursors.


Subject(s)
Anemia/parasitology , Erythropoiesis/physiology , Hemeproteins/pharmacology , Malaria/blood , Reticulocytosis/physiology , Analysis of Variance , Anemia/blood , Anemia/metabolism , Animals , Erythrocyte Count , Erythropoiesis/drug effects , Female , Macrophages/chemistry , Macrophages/parasitology , Malaria/parasitology , Mice , Mice, Inbred C57BL , Monocytes/chemistry , Monocytes/parasitology , Plasmodium , Reticulocytosis/drug effects , Schizonts/physiology
13.
PLoS Pathog ; 9(7): e1003491, 2013.
Article in English | MEDLINE | ID: mdl-23853600

ABSTRACT

Interferon Regulatory Factor 8 (IRF8) is required for development, maturation and expression of anti-microbial defenses of myeloid cells. BXH2 mice harbor a severely hypomorphic allele at Irf8 (Irf8(R294C)) that causes susceptibility to infection with intracellular pathogens including Mycobacterium tuberculosis. We report that BXH2 are completely resistant to the development of cerebral malaria (ECM) following Plasmodium berghei ANKA infection. Comparative transcriptional profiling of brain RNA as well as chromatin immunoprecipitation and high-throughput sequencing (ChIP-seq) was used to identify IRF8-regulated genes whose expression is associated with pathological acute neuroinflammation. Genes increased by infection were strongly enriched for IRF8 binding sites, suggesting that IRF8 acts as a transcriptional activator in inflammatory programs. These lists were enriched for myeloid-specific pathways, including interferon responses, antigen presentation and Th1 polarizing cytokines. We show that inactivation of several of these downstream target genes (including the Irf8 transcription partner Irf1) confers protection against ECM. ECM-resistance in Irf8 and Irf1 mutants is associated with impaired myeloid and lymphoid cells function, including production of IL12p40 and IFNγ. We note strong overlap between genes bound and regulated by IRF8 during ECM and genes regulated in the lungs of M. tuberculosis infected mice. This IRF8-dependent network contains several genes recently identified as risk factors in acute and chronic human inflammatory conditions. We report a common core of IRF8-bound genes forming a critical inflammatory host-response network.


Subject(s)
Brain/immunology , Gene Expression Regulation , Immunity, Innate , Interferon Regulatory Factors/metabolism , Malaria, Cerebral/immunology , Nerve Tissue Proteins/metabolism , Plasmodium berghei/immunology , Amino Acid Substitution , Animals , Binding Sites , Brain/metabolism , Brain/parasitology , Cells, Cultured , Cytokines/biosynthesis , Cytokines/blood , Gene Expression Profiling , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factors/chemistry , Interferon Regulatory Factors/genetics , Malaria, Cerebral/blood , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Mice , Mice, Knockout , Mice, Mutant Strains , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/immunology , Neurons/metabolism , Neurons/parasitology , Specific Pathogen-Free Organisms , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/parasitology
14.
PLoS Negl Trop Dis ; 5(10): e1370, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22039562

ABSTRACT

The murine parasite Heligmosomoides polygyrus is a convenient experimental model to study immune responses and pathology associated with gastrointestinal nematode infections. The excretory-secretory products (ESP) produced by this parasite have potent immunomodulatory activity, but the protein(s) responsible has not been defined. Identification of the protein composition of ESP derived from H. polygyrus and other relevant nematode species has been hampered by the lack of genomic sequence information required for proteomic analysis based on database searches. To overcome this, a transcriptome next generation sequencing (RNA-seq) de novo assembly containing 33,641 transcripts was generated, annotated, and used to interrogate mass spectrometry (MS) data derived from 1D-SDS PAGE and LC-MS/MS analysis of ESP. Using the database generated from the 6 open reading frames deduced from the RNA-seq assembly and conventional identification programs, 209 proteins were identified in ESP including homologues of vitellogenins, retinol- and fatty acid-binding proteins, globins, and the allergen V5/Tpx-1-related family of proteins. Several potential immunomodulators, such as macrophage migration inhibitory factor, cysteine protease inhibitors, galectins, C-type lectins, peroxiredoxin, and glutathione S-transferase, were also identified. Comparative analysis of protein annotations based on the RNA-seq assembly and proteomics revealed processes and proteins that may contribute to the functional specialization of ESP, including proteins involved in signalling pathways and in nutrient transport and/or uptake. Together, these findings provide important information that will help to illuminate molecular, biochemical, and in particular immunomodulatory aspects of host-H. polygyrus biology. In addition, the methods and analyses presented here are applicable to study biochemical and molecular aspects of the host-parasite relationship in species for which sequence information is not available.


Subject(s)
Gene Expression Profiling , Helminth Proteins/analysis , Nematospiroides dubius/chemistry , Nematospiroides dubius/genetics , Proteome/analysis , Animals , Chromatography, Liquid , Electrophoresis, Polyacrylamide Gel , Female , Male , Mice , Mice, Inbred BALB C , Sequence Analysis, DNA , Tandem Mass Spectrometry
15.
J Immunol ; 185(9): 5495-502, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20876354

ABSTRACT

Pathogen sensing by the inflammasome activates inflammatory caspases that mediate inflammation and cell death. Caspase-12 antagonizes the inflammasome and NF-κB and is associated with susceptibility to bacterial sepsis. A single-nucleotide polymorphism (T(125)C) in human Casp12 restricts its expression to Africa, Southeast Asia, and South America. Here, we investigated the role of caspase-12 in the control of parasite replication and pathogenesis in malaria and report that caspase-12 dampened parasite clearance in blood-stage malaria and modulated susceptibility to cerebral malaria. This response was independent of the caspase-1 inflammasome, as casp1(-/-) mice were indistinguishable from wild-type animals in response to malaria, but dependent on enhanced NF-κB activation. Mechanistically, caspase-12 competed with NEMO for association with IκB kinase-α/ß, effectively preventing the formation of the IκB kinase complex and inhibiting downstream transcriptional activation by NF-κB. Systemic inhibition of NF-κB or Ab neutralization of IFN-γ reversed the increased resistance of casp12(-/-) mice to blood-stage malaria infection.


Subject(s)
Caspase 12/immunology , Inflammation/immunology , Malaria/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Animals , Caspase 12/genetics , Cytokines/biosynthesis , Cytokines/immunology , Enzyme Activation/immunology , Enzyme-Linked Immunosorbent Assay , Female , Genetic Predisposition to Disease , Humans , Inflammation/genetics , Malaria/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
16.
Exp Parasitol ; 125(4): 315-24, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20219464

ABSTRACT

In mice, loss of pantetheinase activity causes susceptibility to infection with Plasmodium chabaudi AS. Treatment of mice with the pantetheinase metabolite cysteamine reduces blood-stage replication of P. chabaudi and significantly increases survival. Similarly, a short exposure of Plasmodium to cysteamine ex vivo is sufficient to suppress parasite infectivity in vivo. This effect of cysteamine is specific and not observed with a related thiol (dimercaptosuccinic acid) or with the pantethine precursor of cysteamine. Also, cysteamine does not protect against infection with the parasite Trypanosoma cruzi or the fungal pathogen Candida albicans, suggesting cysteamine acts directly against the parasite and does not modulate host inflammatory response. Cysteamine exposure also blocks replication of P. falciparum in vitro; moreover, these treated parasites show higher levels of intact hemoglobin. This study highlights the in vivo action of cysteamine against Plasmodium and provides further evidence for the involvement of pantetheinase in host response to this infection.


Subject(s)
Antimalarials/pharmacology , Cysteamine/pharmacology , Malaria/drug therapy , Plasmodium chabaudi/drug effects , Plasmodium falciparum/drug effects , Amidohydrolases/metabolism , Animals , Antimalarials/therapeutic use , Candidiasis/drug therapy , Chagas Disease/drug therapy , Chloroquine/pharmacology , Cysteamine/therapeutic use , Cytokines/blood , Cytokines/drug effects , Dose-Response Relationship, Drug , Erythrocytes/drug effects , Erythrocytes/parasitology , Female , GPI-Linked Proteins , Hemoglobins/metabolism , Humans , Malaria/parasitology , Male , Mice , Mice, Inbred C57BL , Parasitemia/drug therapy , Parasitemia/parasitology , Plasmodium falciparum/metabolism , Trypanosoma cruzi/drug effects
17.
Haematologica ; 94(2): 195-204, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19109218

ABSTRACT

BACKGROUND: The contribution of pro-inflammatory cytokines to the pathogenesis of malarial anemia has been studied extensively but the roles of Th2 cytokines remain unknown. Here, we investigated the role of signal transducer and activator of transcription (STAT)6-mediated responses in erythropoietic suppression during acute malaria infection in mice. DESIGN AND METHODS: Naïve and/or erythropoietin-treated wild-type and STAT6(-/-) mice were infected with Plasmodium chabaudi AS (P. chabaudi), and the effects parasitemia, hematologic parameters, erythropoietin receptor, TER119, and CD71 expression, in vitro erythropoietin-stimulated proliferation of splenic erythroid precursors, and serum cytokine levels were analyzed. To explore the role of interleukin-4 in STAT6-dependent erythropoietic suppression, mice were treated in vivo with a monoclonal antibody to interleukin-4 and the effects on parasitemia, hematologic parameters, and cytokine levels were analyzed. RESULTS: Infected STAT6(-/-) mice developed enhanced reticulocytosis compared to wild-type mice despite higher parasitemia and a similar course of anemia. Enhanced reticulocytosis in infected STAT6(-/-) mice was associated with an increased frequency of late-stage erythroblasts, fewer leukocytes expressing CD71, and increased erythropoietin-stimulated proliferation of splenocytes compared to infected wild-type mice. Interleukin-4-depleted wild-type mice had increased levels of parasitemia and a course of reticulocytosis similar to responses observed in infected STAT6(-/-) mice. Determination of serum cytokine levels in STAT6(-/-) and wild-type mice depleted of interleukin-4 by treatment with mAb revealed significantly lower levels of interferon-gamma compared to control wild-type mice during infection. CONCLUSIONS: Together, these findings provide evidence for a STAT6-dependent mechanism in mediating erythropoietic suppression during acute blood-stage malaria and indicate a role for interleukin-4 and possibly interferon-gammain STAT6-induced erythropoietic suppression.


Subject(s)
Anemia/blood , Erythropoiesis , Malaria/blood , STAT6 Transcription Factor/physiology , Animals , Disease Models, Animal , Interferon-gamma , Interleukin-4 , Mice , Th2 Cells/immunology
18.
J Immunol ; 179(4): 2467-76, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17675508

ABSTRACT

The chronic myeloid leukemia syndrome of the BXH-2 mouse strain (Mus musculus) is caused by a recessive mutation (R294C) in the transcriptional regulator Icsbp1/IRF-8. In trans activation assays using an IL-12p40 gene reporter construct introduced in RAW 264.7 mouse macrophages, we show that the Icsbp1(C294) isoform behaves as a partial loss-of-function. The Icsbp1(C294) hypomorph allele appears to have a threshold effect on IL-12 production, with pleiotropic consequences on resistance to different types of infections in vivo. Despite the presence of a resistance Nramp1(G169) allele, BXH-2 mice (Icsbp1(C294)) show impaired control of Mycobacterium bovis (bacille Calmette-Guérin) multiplication both early and late during infection, with uncontrolled replication linked to inability to form granulomas in infected liver and spleen. Studies in informative (BXH-2 x BALB/cJ)F(2) mice show that homozygosity for Icsbp1(C294) causes susceptibility to Salmonella enterica serovar Typhimurium to a level comparable to that seen for mice lacking functional Nramp1 or TLR4. Finally, impaired Icsbp1(C294) function is associated with the following: 1) increased replication of the Plasmodium chabaudi AS malarial parasite during the first burst of blood parasitemia, and 2) recurring waves of high blood parasitemia late during infection. These results show that Icsbp1 is required for orchestrating early innate responses and also long-term immune protection against unrelated intracellular pathogens.


Subject(s)
Immunity, Innate , Interferon Regulatory Factors/immunology , Macrophages/immunology , Malaria/immunology , Mycobacterium bovis/immunology , Salmonella Infections/immunology , Tuberculosis/immunology , Amino Acid Substitution , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/immunology , Cell Line , Genes, Recessive , Immunity, Innate/genetics , Interferon Regulatory Factors/genetics , Interleukin-12 Subunit p40/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Macrophages/microbiology , Macrophages/parasitology , Malaria/genetics , Malaria/veterinary , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Mutation, Missense , Parasitemia/genetics , Parasitemia/immunology , Plasmodium chabaudi/immunology , Salmonella Infections/genetics , Salmonella typhimurium/immunology , Syndrome , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Tuberculosis/genetics , Tuberculosis/veterinary
19.
Blood Cells Mol Dis ; 39(1): 63-9, 2007.
Article in English | MEDLINE | ID: mdl-17466543

ABSTRACT

AcB55, AcB61 and CBA/N-Pk(slc) mice carry loss of function mutations in the erythrocyte specific pyruvate kinase gene (Pklr). In AcB55 and AcB61 (Pklr(I90N)) PK deficiency is protective against blood-stage malaria. The mechanistic basis of protection against malaria is unknown and was studied in these two mutant alleles in vivo. The Pklr(G338D) mutation of the CBA/N-Pk(slc) mutant is shown to be more deleterious than the Pklr(I90N) allele with respect to enzymatic activity and severity of hemolytic anemia, with a more dramatic reduction in the half-life of erythrocytes (increased turnover) in the CBA/N-Pk(slc) mice. The CBA/N-Pk(slc) mice are also shown to be highly resistant to infection with Plasmodium chabaudi AS when compared to CBA/J and CBA/N controls. Resistance to malaria, measured as lower levels of blood-stage replication of P. chabaudi, rapid elimination of infected erythrocytes and increased survival to infection, was greater in the Pklr(G338D) mutant, CBA/N-Pk(slc), than in the Pklr(I90N) mutant strains, AcB55/AcB61. These results strongly suggest a correlation between severity of PK-deficiency and extent of protection against malaria. Additionally, the protective effect is independent of the genetic background on which the Pklr mutations occurred.


Subject(s)
Anemia, Hemolytic, Congenital/genetics , Erythrocytes , Immunity, Innate/genetics , Malaria/genetics , Plasmodium chabaudi , Pyruvate Kinase/deficiency , Anemia, Hemolytic, Congenital/enzymology , Anemia, Hemolytic, Congenital/immunology , Animals , Erythrocytes/enzymology , Erythrocytes/immunology , Erythrocytes/parasitology , Malaria/enzymology , Malaria/immunology , Mice , Mice, Mutant Strains , Plasmodium chabaudi/immunology
20.
J Exp Med ; 204(3): 511-24, 2007 Mar 19.
Article in English | MEDLINE | ID: mdl-17312006

ABSTRACT

Mouse strains AcB55 and AcB61 are resistant to malaria by virtue of a mutation in erythrocyte pyruvate kinase (Pklr(I90N)). Linkage analysis in [AcB55 x A/J] F2 mice detected a second locus (Char9; logarithm of odds = 4.74) that regulates the blood-stage replication of Plasmodium chabaudi AS independently of Pklr. We characterized the 77 genes of the Char9 locus for tissue-specific expression, strain-specific alterations in gene expression, and polymorphic variants that are possibly associated with differential susceptibility. We identified Vnn1/Vnn3 as the likely candidates responsible for Char9. Vnn3/Vnn1 map within a conserved haplotype block and show expression levels that are strictly cis-regulated by this haplotype. The absence of Vnn messenger RNA expression and lack of pantetheinase protein activity in tissues are associated with susceptibility to malaria and are linked to a complex rearrangement in the Vnn3 promoter region. The A/J strain also carries a unique nonsense mutation that leads to a truncated protein. Vanin genes code for a pantetheinase involved in the production of cysteamine, a key regulator of host responses to inflammatory stimuli. Administration of cystamine in vivo partially corrects susceptibility to malaria in A/J mice, as measured by reduced blood parasitemia and decreased mortality. These studies suggest that pantetheinase is critical for the host response to malaria.


Subject(s)
Cell Adhesion Molecules/genetics , Cloning, Molecular , Genetic Predisposition to Disease , Hydrolases/genetics , Malaria/enzymology , Malaria/genetics , Amidohydrolases/deficiency , Amidohydrolases/genetics , Amidohydrolases/physiology , Animals , Chromosome Mapping , GPI-Linked Proteins , Genetic Markers , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Inbred Strains , Plasmodium chabaudi/immunology , Quantitative Trait Loci
SELECTION OF CITATIONS
SEARCH DETAIL
...