Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 191(10): 5160-9, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24133169

ABSTRACT

The mechanism by which protective immunity to Plasmodium is lost in the absence of continued exposure to this parasite has yet to be fully elucidated. It has been recently shown that IFN-γ produced during human and murine acute malaria primes the immune response to TLR agonists. In this study, we investigated whether IFN-γ-induced priming is important to maintain long-term protective immunity against Plasmodium chabaudi AS malaria. On day 60 postinfection, C57BL/6 mice still had chronic parasitemia and efficiently controlled homologous and heterologous (AJ strain) challenge. The spleens of chronic mice showed augmented numbers of effector/effector memory (TEM) CD4(+) cells, which is associated with increased levels of IFN-γ-induced priming (i.e., high expression of IFN-inducible genes and TLR hyperresponsiveness). After parasite elimination, IFN-γ-induced priming was no longer detected and protective immunity to heterologous challenge was mostly lost with >70% mortality. Spontaneously cured mice had high serum levels of parasite-specific IgG, but effector T/TEM cell numbers, parasite-driven CD4(+) T cell proliferation, and IFN-γ production were similar to noninfected controls. Remarkably, the priming of cured mice with low doses of IFN-γ rescued TLR hyperresponsiveness and the capacity to control heterologous challenge, increasing the TEM cell population and restoring the CD4(+) T cell responses to parasites. Contribution of TLR signaling to the CD4(+) T cell responses in chronic mice was supported by data obtained in mice lacking the MyD88 adaptor. These results indicate that IFN-γ-induced priming is required to maintain protective immunity against P. chabaudi and aid in establishing the molecular basis of strain-transcending immunity in human malaria.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Malaria/immunology , Plasmodium chabaudi/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Female , Immunoglobulin G/blood , Lymphocyte Count , Malaria/blood , Malaria/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Parasitemia/immunology , Parasitemia/parasitology , Signal Transduction/immunology
2.
PLoS One ; 6(7): e22434, 2011.
Article in English | MEDLINE | ID: mdl-21814579

ABSTRACT

The pivotal role of spleen CD4(+) T cells in the development of both malaria pathogenesis and protective immunity makes necessary a profound comprehension of the mechanisms involved in their activation and regulation during Plasmodium infection. Herein, we examined in detail the behaviour of non-conventional and conventional splenic CD4(+) T cells during P. chabaudi malaria. We took advantage of the fact that a great proportion of CD4(+) T cells generated in CD1d(-/-) mice are I-A(b)-restricted (conventional cells), while their counterparts in I-A(b-/-) mice are restricted by CD1d and other class IB major histocompatibility complex (MHC) molecules (non-conventional cells). We found that conventional CD4(+) T cells are the main protagonists of the immune response to infection, which develops in two consecutive phases concomitant with acute and chronic parasitaemias. The early phase of the conventional CD4(+) T cell response is intense and short lasting, rapidly providing large amounts of proinflammatory cytokines and helping follicular and marginal zone B cells to secrete polyclonal immunoglobulin. Both TNF-α and IFN-γ production depend mostly on conventional CD4(+) T cells. IFN-γ is produced simultaneously by non-conventional and conventional CD4(+) T cells. The early phase of the response finishes after a week of infection, with the elimination of a large proportion of CD4(+) T cells, which then gives opportunity to the development of acquired immunity. Unexpectedly, the major contribution of CD1d-restricted CD4(+) T cells occurs at the beginning of the second phase of the response, but not earlier, helping both IFN-γ and parasite-specific antibody production. We concluded that conventional CD4(+) T cells have a central role from the onset of P. chabaudi malaria, acting in parallel with non-conventional CD4(+) T cells as a link between innate and acquired immunity. This study contributes to the understanding of malaria immunology and opens a perspective for future studies designed to decipher the molecular mechanisms behind immune responses to Plasmodium infection.


Subject(s)
Antigens, CD1d/physiology , CD4-Positive T-Lymphocytes/immunology , Malaria/immunology , Parasitemia/immunology , Plasmodium chabaudi/immunology , Spleen/immunology , Animals , Antibodies, Protozoan/blood , CD4-Positive T-Lymphocytes/pathology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Interferon-gamma/metabolism , Major Histocompatibility Complex/immunology , Malaria/parasitology , Malaria/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parasitemia/pathology , Spleen/pathology , Tumor Necrosis Factor-alpha/metabolism
3.
J Interferon Cytokine Res ; 30(6): 417-26, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20187775

ABSTRACT

The NK1.1 molecule participates in NK, NKT, and T-cell activation, contributing to IFN-gamma production and cytotoxicity. To characterize the early immune response to Plasmodium chabaudi AS, spleen NK1.1(+) and NK1.1(-) T cells were compared in acutely infected C57BL/6 mice. The first parasitemia peak in C57BL/6 mice correlated with increase in CD4(+)NK1.1(+)TCR-alphabeta(+), CD8(+)NK1.1(+)TCR-alphabeta(+), and CD4(+)NK1.1(-)TCR-alphabeta(+) cell numbers per spleen, where a higher increment was observed for NK1.1(+) T cells compared to NK1.1(-) T cells. According to the ability to recognize the CD1d-alpha-GalCer tetramer, CD4(+)NK1.1(+) cells in 7-day infected mice were not predominantly invariant NKT cells. At that time, nearly all NK1.1(+) T cells and around 30% of NK1.1(-) T cells showed an experienced/activated (CD44(HI)CD69(HI)CD122(HI)) cell phenotype, with high expression of Fas and PD-L1 correlating with their low proliferative capacity. Moreover, whereas IFN-gamma production by CD4(+)NK1.1(+) cells peaked at day 4 p.i., the IFN-gamma response of CD4(+)NK1.1(-) cells continued to increase at day 5 of infection. We also observed, at day 7 p.i., 2-fold higher percentages of perforin(+) cells in CD8(+)NK1.1(+) cells compared to CD8(+)NK1.1(-) cells. These results indicate that spleen NK1.1(+) and NK1.1(-) T cells respond to acute P. chabaudi malaria with different kinetics in terms of activation, proliferation, and IFN-gamma production.


Subject(s)
Malaria/immunology , Natural Killer T-Cells/metabolism , Plasmodium chabaudi/immunology , Spleen/immunology , T-Lymphocyte Subsets/metabolism , Animals , Antigens, CD/biosynthesis , Antigens, Ly/biosynthesis , Cell Proliferation , Immunophenotyping , Interferon-gamma/metabolism , Lymphocyte Activation , Malaria/pathology , Male , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily B/biosynthesis , Natural Killer T-Cells/immunology , Natural Killer T-Cells/parasitology , Natural Killer T-Cells/pathology , Plasmodium chabaudi/pathogenicity , Spleen/parasitology , Spleen/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/parasitology , T-Lymphocyte Subsets/pathology
4.
J Immunol ; 181(12): 8344-55, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19050251

ABSTRACT

The mechanisms responsible for the generation and maintenance of immunological memory to Plasmodium are poorly understood and the reasons why protective immunity in humans is so difficult to achieve and rapidly lost remain a matter for debate. A possible explanation for the difficulty in building up an efficient immune response against this parasite is the massive T cell apoptosis resulting from exposure to high-dose parasite Ag. To determine the immunological mechanisms required for long-term protection against P. chabaudi malaria and the consequences of high and low acute phase parasite loads for acquisition of protective immunity, we performed a detailed analysis of T and B cell compartments over a period of 200 days following untreated and drug-treated infections in female C57BL/6 mice. By comparing several immunological parameters with the capacity to control a secondary parasite challenge, we concluded that loss of full protective immunity is not determined by acute phase parasite load nor by serum levels of specific IgG2a and IgG1 Abs, but appears to be a consequence of the progressive decline in memory T cell response to parasites, which occurs similarly in untreated and drug-treated mice with time after infection. Furthermore, by analyzing adoptive transfer experiments, we confirmed the major role of CD4(+) T cells for guaranteeing long-term full protection against P. chabaudi malaria.


Subject(s)
Antibodies, Protozoan/blood , B-Lymphocyte Subsets/immunology , Immunity, Innate , Immunologic Memory , Malaria/immunology , Malaria/parasitology , Plasmodium chabaudi/immunology , T-Lymphocyte Subsets/immunology , Animals , Antibodies, Protozoan/biosynthesis , B-Lymphocyte Subsets/drug effects , B-Lymphocyte Subsets/parasitology , Erythrocytes/immunology , Erythrocytes/parasitology , Female , Immunity, Innate/drug effects , Immunity, Innate/genetics , Immunologic Memory/drug effects , Immunologic Memory/genetics , Malaria/drug therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Parasitemia/drug therapy , Parasitemia/immunology , Parasitemia/parasitology , Plasmodium chabaudi/drug effects , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/parasitology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...