Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
BMC Immunol ; 11: 56, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21078176

ABSTRACT

BACKGROUND: During the pre-patent stage of infection, juvenile Schistosoma blood flukes co-opt signals from the adaptive immune system to facilitate parasite development, but the types of responses that are induced at this early stage of infection, and the parasite antigens they target, have not been characterized. RESULTS: Through analysis of experimental pre-patent infections, we show that the S. mansoni cysteine protease SmCB1 is rapidly targeted by an antigen-specific IgE response. The induction of this response is independent of schistosome eggs as infection with male or female worms alone also induced SmCB1-specific IgE. We also show that the SmCB1-specific IgE response is dependent on cognate CD4+ T cell help and IL-4, suggesting that pre-patent Th2 responses provide T cell help for the SmCB1-specific IgE response. Finally, exposed human subjects also produced IgE against SmCB1. CONCLUSIONS: Our data demonstrate that, like eggs, schistosome worms also induce functional type 2 responses and that a parasite cysteine protease is an inducer of type 2 responses during the early stages of schistosome infection.


Subject(s)
Cathepsin B/metabolism , Helminth Proteins/metabolism , Interleukin-4/metabolism , Schistosoma mansoni/immunology , Schistosomiasis/immunology , Th2 Cells/immunology , Adaptive Immunity , Animals , Cathepsin B/immunology , Female , Helminth Proteins/immunology , Humans , Immunoglobulin E/blood , Interleukin-4/genetics , Interleukin-4/immunology , Life Cycle Stages , Male , Mice , Schistosoma mansoni/pathogenicity , Schistosomiasis/blood , Th1-Th2 Balance
2.
PLoS Pathog ; 6(4): e1000892, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20442785

ABSTRACT

Schistosoma blood flukes, which infect over 200 million people globally, co-opt CD4+ T cell-dependent mechanisms to facilitate parasite development and egg excretion. The latter requires Th2 responses, while the mechanism underpinning the former has remained obscure. Using mice that are either defective in T cell receptor (TCR) signaling or that lack TCRs that can respond to schistosomes, we show that naïve CD4+ T cells facilitate schistosome development in the absence of T cell receptor signaling. Concurrently, the presence of naïve CD4+ T cells correlates with both steady-state changes in the expression of genes that are critical for the development of monocytes and macrophages and with significant changes in the composition of peripheral mononuclear phagocyte populations. Finally, we show that direct stimulation of the mononuclear phagocyte system restores blood fluke development in the absence of CD4+ T cells. Thus we conclude that schistosomes co-opt innate immune signals to facilitate their development and that the role of CD4+ T cells in this process may be limited to the provision of non-cognate help for mononuclear phagocyte function. Our findings have significance for understanding interactions between schistosomiasis and other co-infections, such as bacterial infections and human immunodeficiency virus infection, which potently stimulate innate responses or interfere with T cell help, respectively. An understanding of immunological factors that either promote or inhibit schistosome development may be valuable in guiding the development of efficacious new therapies and vaccines for schistosomiasis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Schistosoma mansoni/immunology , Schistosomiasis/immunology , Animals , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Gene Expression Profiling , Helminth Proteins/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptors, Antigen, T-Cell/immunology
3.
J Biol Chem ; 285(16): 12063-70, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20164173

ABSTRACT

Toxoplasma gondii, the etiological agent of toxoplasmosis, utilizes stage-specific expression of antigenically distinct glycosylphosphatidylinositol-tethered surface coat proteins to promote and establish chronic infection. Of the three infective stages of T. gondii, sporozoites are encapsulated in highly infectious oocysts that have been linked to large scale outbreaks of toxoplasmosis. SporoSAG (surface antigen glycoprotein) is the dominant surface coat protein expressed on the surface of sporozoites. Using a bioinformatic approach, we show that SporoSAG clusters with the SAG2 subfamily of the SAG1-related superfamily (SRS) and is non-polymorphic among the 11 haplogroups of T. gondii strains. In contrast to the immunodominant SAG1 protein expressed on tachyzoites, SporoSAG is non-immunogenic during natural infection. We report the 1.60 A resolution crystal structure of SporoSAG solved using cadmium single anomalous dispersion. SporoSAG crystallized as a monomer and displays unique features of the SRS beta-sandwich fold relative to SAG1 and BSR4. Intriguingly, the structural diversity is localized to the upper sheets of the beta-sandwich fold and may have important implications for multimerization and host cell ligand recognition. The structure of SporoSAG also reveals an unexpectedly acidic surface that contrasts with the previously determined SAG1 and BSR4 structures where a basic surface is predicted to play a role in binding negatively charged glycosaminoglycans. Our structural and functional characterization of SporoSAG provides a rationale for the evolutionary divergence of this key SRS family member.


Subject(s)
Antigens, Protozoan/chemistry , Protozoan Proteins/chemistry , Toxoplasma/immunology , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , Antigens, Surface/chemistry , Antigens, Surface/genetics , Base Sequence , Crystallography, X-Ray , DNA, Protozoan/genetics , Genes, Protozoan , Humans , Models, Molecular , Phylogeny , Protein Structure, Secondary , Protozoan Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Static Electricity , Toxoplasma/genetics , Toxoplasma/pathogenicity , Toxoplasmosis/immunology , Toxoplasmosis/parasitology
4.
Immunity ; 31(5): 772-86, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19896394

ABSTRACT

Using a model of lethal oral infection with Toxoplasma gondii, we examined the fate of both induced and natural regulatory T (Treg) cells in the face of strong inflammatory responses occurring in a tolerogenic-prone environment. We found that during highly T helper 1 (Th1) cell-polarized mucosal immune responses, Treg cell numbers collapsed via multiple pathways, including blockade of Treg cell induction and disruption of endogenous Treg cell homeostasis. In particular, shutdown of interleukin 2 (IL-2) in the highly Th1 cell-polarized environment triggered by infection directly contributes to Treg cell incapacity to suppress effector responses and eventually leads to immunopathogenesis. Furthermore, we found that environmental cues provided by both local dendritic cells and effector T cells can induce the expression of T-bet transcription factor and IFN-gamma by Treg cells. These data reveal a mechanism for Th1 cell pathogenicity that extends beyond their proinflammatory program to limit Treg cell survival.


Subject(s)
Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , Toxoplasma , Animals , Cell Proliferation , Interleukin-2/metabolism , Mice , Mice, Inbred C57BL , Phenotype , Toxoplasmosis/immunology
5.
Int J Parasitol ; 37(3-4): 405-15, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17196594

ABSTRACT

Schistosoma blood flukes are trematode parasites with a cosmopolitan distribution that infect over 200 million people globally. We previously showed that Schistosoma mansoni growth and development in the mammalian host is dependent on signals from host CD4+ T cells. To gain insight into the mechanisms that underlie this dependence, we sought to determine the evolutionary origins and limits of this aspect of the host-pathogen relationship. By infecting RAG-1-/- mice with a range of different schistosome species and strains, we tested several hypotheses concerning the time during Schistosoma evolution at which this dependence arose, and whether this dependence is specific to Schistosoma or is also found in other blood flukes. Our data indicate that the developmental dependence on CD4+ T cells previously described for S. mansoni is conserved in the evolutionarily basal species Schistosoma japonicum, suggesting this developmental adaptation arose early in Schistosoma evolution. We also demonstrate that the development of the more evolutionarily derived species Schistosoma haematobium and Schistosoma intercalatum are dependent on adaptive immune signals. Together, these data suggest that the blood fluke parasites of humans utilise common mechanisms to infect their hosts and to co-opt immune signals in the coordination of parasite development. Thus, exploitation of host-schistosome interactions to impair or prevent parasite development may represent a novel approach to combating all of the schistosome pathogens of humans.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Schistosoma/growth & development , Schistosomiasis/immunology , Animals , Biological Evolution , Female , Homeodomain Proteins/physiology , Host-Parasite Interactions , Humans , Male , Mice , Mice, Inbred C57BL , Schistosoma/classification , Schistosoma/immunology , Schistosoma haematobium/growth & development , Schistosoma haematobium/immunology , Schistosoma japonicum/growth & development , Schistosoma japonicum/immunology , Schistosomiasis/parasitology , Species Specificity
6.
J Infect Dis ; 194(11): 1609-16, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17083048

ABSTRACT

The human pathogen Schistosoma mansoni exhibits a highly evolved and intricate relationship with its host, evading immune destruction while co-opting CD4(+) T cell-driven mechanisms to facilitate parasite development and egg excretion. Because the common gamma ( gamma (c)) chain cytokine interleukin (IL)-7 is also implicated in modulating schistosome development, we investigated whether this effect is mediated indirectly through the essential role that IL-7 plays in CD4(+) T cell growth and survival. We demonstrate that attenuated schistosome development in the absence of IL-7 results from dysregulated T cell homeostasis and not from disruption of direct interactions between schistosomes and IL-7. We also identify an indirect role that another gamma (c) chain cytokine plays in schistosome development, demonstrating that IL-2 expression by CD4(+) T cells is essential for normal parasite development. Thus, cytokines critical for CD4(+) T cell survival and function can mediate indirect but potent effects on developing schistosomes and underscore the importance of CD4(+) T cells in facilitating schistosome development.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interleukin-2/immunology , Interleukin-7/immunology , Schistosoma mansoni/growth & development , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Adoptive Transfer , Animals , Homeostasis , Liver/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Parasite Egg Count , Receptors, Interleukin-2/genetics , Receptors, Interleukin-7/genetics , Schistosoma mansoni/immunology
7.
Virology ; 353(1): 111-20, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16797051

ABSTRACT

The arenavirus family contains several important human pathogens including Lassa fever virus (LASV), lymphocytic choriomeningitis virus (LCMV) and the New World clade B viruses Junin (JUNV) and Machupo (MACV). Previously, alpha-dystroglycan (alpha-DG) was identified as a receptor recognized by LASV and certain strains of LCMV. However, other studies have suggested that alpha-DG is probably not used by the clade B viruses, and the receptor(s) for these pathogens is currently unknown. Using pseudotyped retroviral vectors displaying arenavirus glycoproteins (GPs), we are able to explore the role played by the GP in viral entry in the absence of other viral proteins. By examining the ability of the vectors to transduce DG knockout murine embryonic stem (ES) cells, we have confirmed that LASV has an absolute requirement for alpha-DG in these cells. However, the LCMV GP can still direct substantial entry into murine ES cells in the absence of alpha-DG, even when the GP from the clone 13 variant is used that has previously been reported to be highly dependent on alpha-DG for entry. We also found that neither LASV or LCMV pseudotyped vectors were able to transduce human or murine lymphocytes, presumably due to the glycosylation state of alpha-DG in these cells. In contrast, the JUNV and MACV GPs displayed broad tropism on human, murine and avian cell types, including lymphocytes, and showed no requirement for alpha-DG in murine ES cells. These findings highlight the importance of molecules other than alpha-DG for arenavirus entry. An alternate receptor is present on murine ES cells that can be used by LCMV but not by LASV, and which is not available on human or murine lymphocytes, while a distinct and widely expressed receptor(s) is used by the clade B viruses.


Subject(s)
Arenaviridae/pathogenicity , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism , Stem Cells/cytology , Animals , Arenaviridae/classification , CHO Cells , COS Cells , Cell Culture Techniques , Cell Line , Chlorocebus aethiops , Cricetinae , HeLa Cells , Humans , Jurkat Cells , Mice , NIH 3T3 Cells , Vero Cells
8.
Exp Biol Med (Maywood) ; 230(9): 659-67, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16179734

ABSTRACT

The flaky skin (fsn) mutation in mice causes pleiotropic abnormalities including psoriasiform dermatitis, anemia, hyper-IgE, and anti-dsDNA autoantibodies resembling those detected in systemic lupus erythematosus. The fsn mutation was mapped to an interval of 3.9 kb on chromosome 17 between D17Mit130 and D17Mit162. Resequencing of known and predicted exons and regulatory sequences from this region in fsn/fsn and wild-type mice indicated that the mutation is due to the insertion of an endogenous retrovirus (early transposon class) into intron 14 of the Tetratricopeptide repeat (TPR) domain 7 (Ttc7) gene. The insertion leads to reduced levels of wild-type Ttc7 transcripts in fsn mice and the insertion of an additional exon derived from the retrovirus into the majority of Ttc7 mRNAs. This disrupts one of the TPRs within TTC7 and may affect its interaction with an as-yet unidentified protein partner. The Ttc7 is expressed in multiple types of tissue including skin, kidney, spleen, and thymus, but is most abundant in germinal center B cells and hematopoietic stem cells, suggesting an important role in the development of immune system cells. Its role in immunologic and hematologic disorders should be further investigated.


Subject(s)
Anemia/genetics , Autoimmunity/genetics , Mutation , Proteins/genetics , Psoriasis/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Gene Expression , Genetic Linkage , Humans , Lymphocytes/immunology , Mice , Mice, Inbred Strains , Molecular Sequence Data , Phenotype , Sequence Homology, Amino Acid
9.
Immunol Rev ; 201: 57-74, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15361233

ABSTRACT

Components of the type 2 immune response may mediate host protection against both helminthic parasites and harmful allergic responses. A central player in this response is the T-helper 2 (Th2) effector cell, which produces interleukin (IL)-4, IL-5, IL-13, and other Th2 cytokines during the primary and memory response. Specific aspects of the parasite that trigger Th2-cell differentiation are not yet defined. Furthermore, the cell types and cell surface and secreted molecules that provide the immune milieu required for the development of Th2 effector cells and also Th2 memory cells are not well understood. They will probably vary with the particular helminth or other antigen inducing the Th2 response. We have used third stage larvae of intestinal nematode parasites as adjuvants to promote naïve nonparasite antigen-specific T cells to differentiate into Th2 cells. This model system avoids possible parasite antigen-specific T-cell clones or cross-reactive memory T cells that may preferentially differentiate into Th2 effector cells during the course of infection and confound the stereotypical components of parasite-induced Th2 cell differentiation. We have found that these parasites have a potent adjuvant effect and have used our model system to begin to investigate the events that lead to the development of polarized Th2 cells in vivo.


Subject(s)
Interleukin-4/biosynthesis , Intestinal Diseases, Parasitic/immunology , Strongylida Infections/immunology , Strongylida/immunology , Th2 Cells/immunology , Animals , Intestinal Diseases, Parasitic/parasitology , Mice , Mice, Inbred BALB C , Strongylida Infections/parasitology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL