Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Science ; 345(6196): 573-7, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24968940

ABSTRACT

Mammals are coinfected by multiple pathogens that interact through unknown mechanisms. We found that helminth infection, characterized by the induction of the cytokine interleukin-4 (IL-4) and the activation of the transcription factor Stat6, reactivated murine γ-herpesvirus infection in vivo. IL-4 promoted viral replication and blocked the antiviral effects of interferon-γ (IFNγ) by inducing Stat6 binding to the promoter for an important viral transcriptional transactivator. IL-4 also reactivated human Kaposi's sarcoma-associated herpesvirus from latency in cultured cells. Exogenous IL-4 plus blockade of IFNγ reactivated latent murine γ-herpesvirus infection in vivo, suggesting a "two-signal" model for viral reactivation. Thus, chronic herpesvirus infection, a component of the mammalian virome, is regulated by the counterpoised actions of multiple cytokines on viral promoters that have evolved to sense host immune status.


Subject(s)
Gammaherpesvirinae/physiology , Herpesvirus 8, Human/physiology , Interferon-gamma/immunology , Interleukin-4/metabolism , STAT6 Transcription Factor/metabolism , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Virus Activation/physiology , Animals , Gammaherpesvirinae/genetics , Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Humans , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Nematospiroides dubius/immunology , Ovum/immunology , Promoter Regions, Genetic , Strongylida Infections/immunology , Virus Activation/drug effects , Virus Activation/genetics , Virus Latency/physiology , Virus Replication/physiology
2.
Parasite Immunol ; 35(9-10): 248-55, 2013.
Article in English | MEDLINE | ID: mdl-23841670

ABSTRACT

Recent advances in molecular genetics and in imaging mean that it is now increasingly feasible to image biological processes within helminth parasites and to visualize interactions between worms and their hosts. Moreover, other innovative imaging approaches that are not dependent on transgenic parasites have been applied to, and or developed for, the study of helminth parasites and have provided novel and important insights into the biology of these important pathogens.


Subject(s)
Nematoda/cytology , Nematode Infections , Trematoda/cytology , Trematode Infections , Animals , Humans , Microscopy, Electron , Nematoda/physiology , Nematode Infections/parasitology , Trematoda/physiology , Trematode Infections/parasitology
3.
Parasite Immunol ; 31(12): 741-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19891612

ABSTRACT

Protection against Mesocestoides corti, a cestode that invades vital organs, is dependent on the production of IL-4, as IL-4(-/-) mice were found to have higher parasite burdens when compared with wild-type mice. The goal of this study was to investigate the role of IL-4 in immunity to M. corti, focusing on the immunological profile and on potential mediators of pathology. IL-4(-/-) mice infected with M. corti showed 100% mortality by 32 days, whereas wild-type mice survived for approximately 1 year. Parasite burdens were significantly increased in the liver, peritoneal, and thoracic cavities of IL-4(-/-) mice, associated with impaired recruitment of inflammatory cells and a reduction in monocytes and macrophages. IL-5 production by splenocytes and expression in liver tissue was decreased in infected IL-4(-/-) mice compared with wild-type mice. In contrast, IL-4(-/-) mice produced increased amounts of IFNgamma and TNFalpha. Alternatively activated macrophages were a major feature of liver granulomas in wild-type mice evidenced by Arginase I expression, while livers from infected IL-4(-/-) mice showed impaired alternative macrophage activation without increased classical macrophage activation. Thus, lethality during M. corti infection of IL-4(-/-) mice is associated with decreased Th2 cytokines, increased Th1 cytokines and impairment of alternatively activated macrophages.


Subject(s)
Cestode Infections/immunology , Host-Parasite Interactions/physiology , Interleukin-4/immunology , Macrophages/immunology , Mesocestoides , Th2 Cells/immunology , Animals , Cestode Infections/metabolism , Cestode Infections/parasitology , Host-Parasite Interactions/immunology , Interferon-gamma/biosynthesis , Interleukin-4/genetics , Interleukin-5/biosynthesis , Liver/immunology , Liver/parasitology , Macrophage Activation , Mice , Mice, Inbred BALB C , Mice, Knockout , Spleen/immunology , Spleen/metabolism , Th2 Cells/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
4.
Parasite Immunol ; 30(4): 215-21, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18324924

ABSTRACT

The sequencing of the schistosome genome and the establishment of techniques for RNAi and transient transfection in these parasites have opened the door for a reverse genetics approach to studying schistosomes. One of the most intriguing aspects of schistosome biology is the interaction of these parasites with the immune system. The immune response underlies the ability of the host to survive while infected and to eventually develop resistance to further infection. However, it is also instrumental in the development of disease due to its role orchestrating granuloma formation around tissue-trapped parasite eggs. While schistosomes have clearly evolved mechanisms for evading host immune responses, their normal development is, paradoxically, also dependent upon the presence of a normal immune system. This article will review recent advances in the development of tools for studying gene function in schistosomes, and discuss how these new tools may be exploited to investigate issues of key importance in the interaction of schistosomes with the host immune system.


Subject(s)
Host-Parasite Interactions , Molecular Biology/methods , Schistosoma/genetics , Schistosoma/immunology , Animals , Schistosomiasis/immunology , Schistosomiasis/transmission
5.
Parasite Immunol ; 27(7-8): 265-70, 2005.
Article in English | MEDLINE | ID: mdl-16138847

ABSTRACT

Schistosomiasis mansoni is a chronic disease caused by infection with helminths of the genus Schistosoma mansoni. Adult schistosomes live intravascularly, and for transmission of this infection it is necessary for parasite eggs to traverse the endothelium, and migrate to the intestinal lumen, from where they can exit the body to continue the lifecycle. This process is dependent on an intact host CD4 T helper (Th) cell response to egg antigens. Perhaps because of this, eggs have evolved to be highly immunogenic and capable of inducing potent Th responses. The egg-induced Th response is unusual in that it is highly Th2-polarized. The selective pressure on the host to mount a Th2 response against eggs is apparent in the fact that Th2 response-defective mice develop acutely lethal disease when infected with schistosomes. In this review I will focus on the underlying basis for the Th2 bias in the immune response to egg antigens.


Subject(s)
Ovum/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Animals , Antigens, Helminth/immunology , Dendritic Cells/immunology , Humans , Mice , Schistosoma mansoni/growth & development , Th2 Cells/immunology
6.
J Immunol ; 167(11): 6533-44, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11714822

ABSTRACT

Type 2 cytokines regulate fibrotic liver pathology in mice infected with Schistosoma mansoni. Switching the immune response to a type 1-dominant reaction has proven highly effective at reducing the pathologic response. Activation of NOS-2 is critical, because type 1-deviated/NO synthase 2 (NOS-2)-deficient mice completely fail to control their response. Here, we demonstrate the differential regulation of NOS-2 and arginase type 1 (Arg-1) by type 1/type 2 cytokines in vivo and for the first time show a critical role for arginase in the pathogenesis of schistosomiasis. Using cytokine-deficient mice and two granuloma models, we show that induction of Arg-1 is type 2 cytokine dependent. Schistosome eggs induce Arg-1, while Mycobacterium avium-infected mice develop a dominant NOS-2 response. IFN-gamma suppresses Arg-1 activity, because type 1 polarized IL-4/IL-10-deficient, IL-4/IL-13-deficient, and egg/IL-12-sensitized animals fail to up-regulate Arg-1 following egg exposure. Notably, granuloma size decreases in these type-1-deviated/Arg-1-unresponsive mice, suggesting an important regulatory role for Arg-1 in schistosome egg-induced pathology. To test this hypothesis, we administered difluoromethylornithine to block ornithine-aminodecarboxylase, which uses the product of arginine metabolism, L-ornithine, to generate polyamines. Strikingly, granuloma size and hepatic fibrosis increased in the ornithine-aminodecarboxylase-inhibited mice. Furthermore, we show that type 2 cytokine-stimulated macrophages produce proline under strict arginase control. Together, these data reveal an important regulatory role for the arginase biosynthetic pathway in the regulation of inflammation and demonstrate that differential activation of Arg-1/NOS-2 is a critical determinant in the pathogenesis of granuloma formation.


Subject(s)
Arginase/metabolism , Arginine/metabolism , Granuloma/immunology , Granuloma/pathology , Nitric Oxide Synthase/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Arginase/antagonists & inhibitors , Arginase/biosynthesis , Cells, Cultured , Disease Models, Animal , Eflornithine/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Activation/immunology , Enzyme Induction/drug effects , Enzyme Induction/genetics , Enzyme Induction/immunology , Enzyme Inhibitors/pharmacology , Female , Granuloma/enzymology , Granuloma/prevention & control , Interleukin-12/physiology , Liver/enzymology , Liver Cirrhosis/enzymology , Liver Cirrhosis/pathology , Lung Diseases, Parasitic/enzymology , Lung Diseases, Parasitic/genetics , Lung Diseases, Parasitic/immunology , Lung Diseases, Parasitic/pathology , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium avium/immunology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Ornithine Decarboxylase Inhibitors , Ovum/immunology , Proline/biosynthesis , Schistosomiasis mansoni/enzymology , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/pathology , Th1 Cells/enzymology , Th2 Cells/enzymology , Tuberculosis/enzymology , Tuberculosis/genetics , Tuberculosis/immunology , Tuberculosis/pathology , Up-Regulation/genetics , Up-Regulation/immunology
7.
Infect Immun ; 69(12): 7445-52, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11705919

ABSTRACT

In the absence of interleukin-4 (IL-4), infection with Schistosoma mansoni leads to a severe fatal disease rather than the chronic survivable condition that occurs in wild-type (WT) mice. Because the sustained production of NO most closely correlates to weight loss and fatality in infected IL-4(-/-) mice and because gamma interferon (IFN-gamma) is an important inducer of inducible NO synthase, infected IL-4(-/-) mice were treated with anti-IFN-gamma antibodies to determine the role of IFN-gamma during schistosomiasis in WT and IL-4(-/-) animals. When IFN-gamma was neutralized, Th2 responses were enhanced and NO production was reduced in both WT and IL-4(-/-) mice. The decreased NO production correlated with a rescue of proliferation in splenocytes from infected IL-4(-/-) mice. Furthermore, the neutralization of IFN-gamma in vivo improved the gross appearance of the liver and led to a reduction in granuloma size in infected IL-4(-/-) but not WT mice. However, the neutralization of IFN-gamma in vivo did not affect the development of severe disease in infected IL-4(-/-) mice. These results suggest that while the increased production of IFN-gamma does lead to some of the pathology observed in infected IL-4(-/-) mice, it is not ultimately responsible for cachexia and death.


Subject(s)
Interferon-gamma/immunology , Interleukin-4/deficiency , Schistosomiasis mansoni/etiology , Animals , Antibodies, Monoclonal , Antigens, Helminth , Cell Division , Female , Interferon-gamma/antagonists & inhibitors , Interleukin-4/genetics , Liver/pathology , Mice , Mice, Mutant Strains , Neutralization Tests , Nitric Oxide/biosynthesis , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/mortality , Spleen/cytology , Spleen/immunology , Th2 Cells
8.
J Leukoc Biol ; 70(5): 737-44, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11698493

ABSTRACT

T cell responses of interleukin (IL)-4(-/-) and wild-type (WT) mice infected with the helper T cell 2 (Th2) response-inducing pathogen Schistosoma mansoni were compared. As expected, given the important role of IL-4 in Th2 response induction, the absence of IL-4 resulted in diminished Th2 responses, apparent as reduced production of IL-4, -5, and -10 by CD4(+) cells isolated from the spleens of infected IL-4(-/-) mice. Surprisingly, these cells produced significantly less interferon (IFN)-gamma and proliferated less than did those from infected WT mice after T cell receptor ligation. CD8(+) cells isolated from infected IL-4(-/-) mice also produced less IFN-gamma than WT CD8 cells, although there was no difference in the proliferative responses of these cell populations. After infection, spleens of infected IL-4(-/-) mice did not enlarge to the same extent as those of WT mice, and attrition of the CD8(+) cell population within this lymphoid organ was noted. Taken together, the data indicate that in addition to inhibiting Th2 response development, the lack of IL-4 during schistosomiasis significantly affects additional aspects of T cell responses.


Subject(s)
Interleukin-4/physiology , Lymphokines/metabolism , Schistosomiasis mansoni/immunology , T-Lymphocyte Subsets/immunology , Animals , Apoptosis , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Progression , Female , Hyperplasia , Immunologic Deficiency Syndromes/etiology , Immunologic Deficiency Syndromes/immunology , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-13/metabolism , Interleukin-2/pharmacology , Interleukin-4/deficiency , Interleukin-4/genetics , Interleukin-5/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromonab-CD3/pharmacology , Schistosomiasis mansoni/pathology , Spleen/pathology , Splenomegaly/etiology , T-Lymphocyte Subsets/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
9.
J Immunol ; 167(4): 1982-8, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11489979

ABSTRACT

Whether dendritic cells (DC) play a passive or active role in Th2 response induction is poorly understood. In this study, we show that CD8- DC pulsed with Th2-polarizing Ag (soluble egg Ag (SEA)) from Schistosoma mansoni potently stimulate Th2 responses in vivo and in vitro while failing to undergo a conventional maturation process. Thus, in contrast to DC pulsed with the Th1 response inducing Ag Propionebacterium acnes, SEA-exposed DC exhibit a phenotype that is most similar to that of immature DC, failing to up-regulate expression of CD40, CD54, CD80, CD86, or OX40L; producing no detectable IL-4, IL-10, or IL-12; and displaying only a minor increase in MHC class II expression. Importantly, in vitro derived DC exposed to SEA were phenotypically similar to CD8- DC isolated from active S. mansoni infection. By discriminating between different types of pathogen and responding appropriately, CD8- DC play a major role in the decision process to mount either a Th1 or Th2 response.


Subject(s)
CD8 Antigens , Dendritic Cells/immunology , Th2 Cells/immunology , Animals , Antigens, Bacterial/pharmacology , Antigens, Helminth/pharmacology , CD40 Antigens/physiology , CD8 Antigens/biosynthesis , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Dendritic Cells/parasitology , Female , Immunophenotyping , Interleukin-12/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Propionibacterium acnes/immunology , Schistosoma mansoni/immunology , Th1 Cells/immunology
10.
J Biol Chem ; 276(34): 31613-9, 2001 Aug 24.
Article in English | MEDLINE | ID: mdl-11406634

ABSTRACT

The biology of the helminth parasite Schistosoma mansoni is closely integrated with that of its mammalian host. SmRK1, a divergent type I transforming growth factor-beta (TGF-beta) receptor of unknown ligand specificity, was previously identified as a candidate for a receptor that allows schistosomes to respond to host-derived growth factors. The TGF-beta family includes activin, bone morphogenetic proteins (BMPs), and TGF-beta, all of which can play crucial roles in metazoan development. The downstream signaling protein of receptors that respond to TGF-beta and activin is Smad2, whereas the receptors that respond to BMPs signal via Smad1. When a constitutively active mutant of SmRK1 was overexpressed with either schistosome Smad1 (SmSmad1) or SmSmad2, a receptor-dependent modulation of SmSmad phosphorylation and luciferase reporter activity occurred only with SmSmad2. To evaluate potential ligand activators of SmRK1, a chimeric receptor containing the extracellular domain of SmRK1 joined to the intracellular domain of the human type I TGF-beta receptor was used. The chimeric receptor bound radiolabeled TGF-beta and could activate a luciferase reporter gene in response to both TGF-beta 1 and TGF-beta 3 but not BMP7. Confirmatory results were obtained using full-length SmRK1. These experiments implicate TGF-beta as a ligand for SmRK1 and as a potential host-derived regulator of parasite growth and development.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Schistosoma mansoni/enzymology , Transforming Growth Factor beta/physiology , Amino Acid Sequence , Animals , Enzyme Activation , Humans , Ligands , Mutation , Precipitin Tests , Protein Binding , Receptors, Transforming Growth Factor beta/chemistry , Sequence Homology, Amino Acid , Signal Transduction/physiology
12.
FEBS Lett ; 490(1-2): 65-9, 2001 Feb 09.
Article in English | MEDLINE | ID: mdl-11172812

ABSTRACT

Schistosoma mansoni receptor kinase-1 (SmRK1) is a divergent type I transforming growth factor beta (TGFbeta) receptor on the surface of adult parasites. Using the intracellular domain of SmRK1 as bait in a yeast two-hybrid screen we identified an interaction with S. mansoni 14-3-3epsilon. The interaction which is phosphorylation-dependent is not specific to schistosomes since 14-3-3epsilon also binds to TbetaRI, the human type I TGFbeta receptor. 14-3-3epsilon enhances TGFbeta-mediated signaling by TbetaRI and is the first TbetaRI-interacting non-Smad protein identified that positively regulates this receptor. The interaction of 14-3-3epsilon with schistosome and human TbetaRI suggests a conserved, but previously unappreciated, role for this protein in TGFbeta signaling pathways.


Subject(s)
Activin Receptors, Type I , Helminth Proteins , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Tyrosine 3-Monooxygenase/physiology , 14-3-3 Proteins , Amino Acid Sequence , Animals , COS Cells , DNA, Complementary/metabolism , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Molecular Sequence Data , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Cell Surface/metabolism , Schistosoma/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Smad Proteins , Trans-Activators/metabolism , Two-Hybrid System Techniques
13.
Infect Immun ; 69(3): 1454-62, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179312

ABSTRACT

We previously demonstrated that mice concurrently infected with Schistosoma mansoni and Toxoplasma gondii undergo accelerated mortality which is preceded by severe liver damage. Abnormally high levels of serum tumor necrosis factor alpha (TNF-alpha) in the dually infected mice suggested a role for this and related proinflammatory mediators in the pathologic alterations. In order to evaluate the factors involved in increased inflammatory-mediator production and mortality, interleukin-12(-/-) (IL-12(-/-)) mice were coinfected with S. mansoni and T. gondii, and survival and immune responses were monitored. These IL-12(-/-) mice displayed decreased liver damage and prolonged time to death relative to wild-type animals also coinfected with these parasites. Relative to the response of cells from the coinfected wild-type animals, levels of TNF-alpha, gamma interferon, and NO produced by splenocytes from coinfected IL-12(-/-) mice were reduced, and levels of IL-5 and IL-10 were increased, with the net result that the immune response of the dually infected IL-12(-/-) mice was similar to that of the wild-type mice infected with S. mansoni alone. While dually infected wild-type animals succumb in the absence of overt parasitemia, the delayed death in the absence of IL-12 is associated with relatively uncontrolled T. gondii replication. These data support the view that S. mansoni-infected mice are acutely sensitive to infection with T. gondii as a result of their increased hepatic sensitivity to high levels of proinflammatory cytokines; IL-12 and TNF-alpha are implicated in this process.


Subject(s)
Interleukin-12/immunology , Liver/pathology , Schistosomiasis mansoni/complications , Toxoplasmosis, Animal/complications , Animals , Female , Inflammation Mediators/metabolism , Interleukin-12/genetics , Mice , Mice, Mutant Strains , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/mortality , Schistosomiasis mansoni/pathology , Survival Analysis , Survivors , Th2 Cells , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Animal/mortality , Toxoplasmosis, Animal/pathology
14.
J Immunol ; 166(3): 1903-11, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11160238

ABSTRACT

Liver enlargement and hepatocyte proliferation, normal responses in wild-type (WT) mice infected with the parasitic helminth Schistosoma mansoni, were found to be severely impaired in infected IL-4(-/-) mice. Compared with WT mice, increased levels of O(2)(-), NO, and the more highly reactive ONOO(-) were detected in the liver and produced by lesional cells isolated from liver granulomas of infected IL-4(-/-) mice. Concurrently, antioxidant defenses in the liver, specifically catalase levels, diminished dramatically during the course of infection in these animals. This contrasted to the situation in infected WT mice, where catalase levels remained as high as those in normal mice. Actual levels of reactive oxygen and nitrogen intermediates in the livers of infected IL-4(-/-) animals are thus likely to be considerably higher than those in the livers of infected WT mice. To determine whether these changes contributed to the development of the more severe disease that characterizes infection in the IL-4(-/-) animals, we treated infected IL-4(-/-) mice with uric acid, a potent scavenger of ONOO(-). This resulted in significantly increased hepatocyte proliferation, decreased morbidity, and prolonged survival. Taken together, these data indicate that IL-4 is playing a protective role during schistosomiasis by controlling the tight regulation of the generation of reactive oxygen and nitrogen intermediates in the liver.


Subject(s)
Interleukin-4/physiology , Liver/immunology , Liver/metabolism , Oxidative Stress/immunology , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/metabolism , Acute Disease , Animals , Antioxidants/administration & dosage , Catalase/antagonists & inhibitors , Catalase/biosynthesis , Cell Division/genetics , Cell Division/immunology , Female , Granuloma/enzymology , Granuloma/genetics , Granuloma/metabolism , Hepatocytes/pathology , Injections, Intraperitoneal , Interleukin-4/deficiency , Interleukin-4/genetics , Liver/enzymology , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Nitrogen/metabolism , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism , Schistosomiasis mansoni/mortality , Schistosomiasis mansoni/pathology , Survival Rate , Uric Acid/administration & dosage
15.
Infect Immun ; 69(1): 589-92, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11119559

ABSTRACT

An interleukin-4 (IL-4)-dependent Th2 response allows wild-type mice to survive infection with the parasite Schistosoma mansoni. In the absence of IL-4, infected mice mount a Th1-like proinflammatory response, develop severe disease, and succumb. Neither the Th1 response nor morbidity is IL-12 dependent in this system.


Subject(s)
Interleukin-12/physiology , Interleukin-4/physiology , Schistosomiasis mansoni/immunology , Animals , Interferon-gamma/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/physiology , Tumor Necrosis Factor-alpha/physiology
16.
Mol Biochem Parasitol ; 111(1): 131-42, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11087923

ABSTRACT

To begin to understand the molecular basis of communication between the parasite Schistosoma mansoni and its mammalian host, we are studying the signaling pathway downstream of S. mansoni receptor kinase-1 (SmRK1), a divergent type I transforming growth factor-beta (TGF-beta) receptor found on the tegumental surface of the parasite. In this study, we have used a homology based PCR approach to clone two S. mansoni Smad (SmSmad) genes; Smads play a pivotal role in the most well understood signaling pathways initiated by the TGF-beta family of ligands in other organisms. Comparison of the amino acid sequences with those of other Smads reveals that the conserved MH1 and MH2 domains of SmSmads show a high degree of identity to homologues in Drosophila. Transcripts for both SmSmads are detected in the same developmental stages as SmRK1, and both are capable of interacting with the intracellular domain of the receptor in vitro. Functional characterization using the human type I TGF-beta receptor further confirms the highly conserved nature of these proteins, as both SmSmads show TGF-beta dependent enhancement of luciferase activity and nuclear translocation in mammalian cells. These data are the first to show a TGF-beta-like receptor/Smad signaling pathway in parasitic helminths and by analogy with other systems, is likely important in regulating schistosome development.


Subject(s)
Activin Receptors, Type I , DNA-Binding Proteins/metabolism , Helminth Proteins , Protein Serine-Threonine Kinases/metabolism , Receptors, Cell Surface/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Schistosoma mansoni/metabolism , Signal Transduction , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Nucleus/metabolism , Cloning, Molecular , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genes, Reporter , Ligands , Luciferases/genetics , Molecular Sequence Data , Polymerase Chain Reaction , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Recombinant Fusion Proteins/metabolism , Schistosoma mansoni/genetics , Schistosoma mansoni/growth & development , Sequence Alignment , Smad Proteins , Smad2 Protein , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription, Genetic , Transfection
17.
J Immunol ; 164(5): 2419-26, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10679078

ABSTRACT

The eggs of Schistosoma mansoni are strong inducers of a Th2 response, and previous work has shown that Ag-specific IL-6 is produced within 24 h after the injection of eggs into mice. Investigations to determine the role of IL-6 in orchestrating the early response to schistosome eggs have revealed that IL-12 is rapidly produced in lymph node cell cultures from egg-injected mice. This "early" IL-12 primes for the production of IL-6 and IFN-gamma, for in IL-12-/- mice egg injection fails to stimulate increased production of either of these cytokines. Furthermore, IL-6 also up-regulates IL-10 production which, together with IL-6, negatively regulates IL-12 and IFN-gamma production. Finally, IL-10 down-regulates the production of its inducer, IL-6. These data indicate that the anti-inflammatory role of IL-6 may be effected through negative regulation of type 1 (IFN-gamma) and type 1-associated (IL-12) cytokines either directly (by IL-6) or indirectly (through the induction of IL-10) and suggest that one mechanism by which eggs may support the development of Th2 responses is through the negative regulation of the type 1 response.


Subject(s)
Antigens, Helminth/immunology , Interleukin-6/physiology , Ovum/immunology , Schistosoma mansoni/immunology , Animals , Antigens, Helminth/administration & dosage , Cells, Cultured , Down-Regulation/immunology , Drug Synergism , Female , Injections, Subcutaneous , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-12/antagonists & inhibitors , Interleukin-12/biosynthesis , Interleukin-12/physiology , Interleukin-6/biosynthesis , Interleukin-6/deficiency , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
18.
Mol Biochem Parasitol ; 104(2): 299-310, 1999 Nov 30.
Article in English | MEDLINE | ID: mdl-10593183

ABSTRACT

The surface membrane of the intravascular parasite Schistosoma mansoni is composed of not one but two closely apposed lipid bilayers which overlie a syncytial cellular layer, known as the tegument or neodermis. To gain insights into how membrane proteins are transported to and displayed on this unusual surface structure, we have investigated the post-translational modification and targeting of SmRK-1, a receptor and type I membrane protein expressed on the parasite surface, using heterologous expression systems. While SmRK-1 enters the secretory pathway in these systems, our data indicate that the SmRK-1 N-terminal signal peptide is either not cleaved by signal peptidase or is only eleven amino acids long or less. Retention of the signal peptide is accompanied by N-linked glycosylation of an asparagine residue within the predicted signal peptide. The SmRK-1 signal peptide is not capable of directing another cytoplasmic protein to the secretory pathway, suggesting that the signal for insertion of the SmRK-1 extracellular domain into the endoplasmic reticulum resides elsewhere in the protein. Further, SmRK-1 is inefficiently transported to the cell surface in mammalian cells, suggesting that the schistosome neodermis possesses specialized systems for receptor targeting and localization.


Subject(s)
Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Schistosoma mansoni/metabolism , Animals , Baculoviridae/genetics , COS Cells , Cell Line , Glycosylation , Helminth Proteins/genetics , Helminth Proteins/metabolism , Insecta , Protein Biosynthesis , Protein Sorting Signals , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Schistosoma mansoni/genetics , Transcription, Genetic
19.
Mol Biochem Parasitol ; 104(2): 285-97, 1999 Nov 30.
Article in English | MEDLINE | ID: mdl-10593182

ABSTRACT

Specialized regions of cellular membranes termed detergent-insoluble glycosphingolipid-enriched membrane domains (DIG) have been identified in mammalian cells and shown to contain signalling molecules, cholesterol, sphingolipids and caveolae. Here we report that the unusual double surface membrane of the tegument of the trematode parasite Schistosoma mansoni possesses biochemically distinct domains analogous to DIG. When subjected to sucrose density gradient centrifugation, a detergent-extracted tegument from adult parasites yielded a low-density fraction consisting of detergent-insoluble complexes (DIC). Several tegument proteins were concentrated in DIC and a subset of these were labelled when adult schistosomes were biotinylated using a membrane-impermeant reactive biotin prior to extraction. The GPI-linked proteins alkaline phosphatase (SmAP), Sm200, the membrane-bound protein Sm23, and a protein recognized by an antibody against human caveolin, co-purified with DIC whereas soluble proteins, such as paramyosin and aldolase, were found at the bottom of the gradient. Antibodies against DIC immunoprecipitated a subset of worm surface proteins and immunolabeled the dorsal tegument of adult worms. Transmission electron microscopy of DIC revealed caveolae-like structures in the double bilayer surface structure. These results suggest that the tegument of adult S. mansoni possesses specialized membrane domains that are resistant to detergent-extraction, contain a subset of total tegument membrane proteins, and bear caveola-like invaginations, and thus are analogous to DIG.


Subject(s)
Caveolins , Helminth Proteins/isolation & purification , Membrane Proteins/isolation & purification , Schistosoma mansoni/ultrastructure , Animals , Blotting, Western , Caveolin 1 , Centrifugation, Density Gradient , Detergents , Helminth Proteins/chemistry , Helminth Proteins/immunology , Membrane Proteins/chemistry , Membrane Proteins/immunology , Membranes/chemistry , Membranes/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Electron , Microscopy, Fluorescence , Schistosoma mansoni/chemistry , Schistosoma mansoni/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...