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.
J Immunol ; 199(2): 613-623, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28584007

ABSTRACT

Tuberculosis (TB), caused by Mycobacterium tuberculosis infection, is a leading cause of mortality and morbidity, causing ∼1.5 million deaths annually. CD4+ T cells and several cytokines, such as the Th1 cytokine IFN-γ, are critical in the control of this infection. Conversely, the immunosuppressive cytokine IL-10 has been shown to dampen Th1 cell responses to M. tuberculosis infection impairing bacterial clearance. However, the critical cellular source of IL-10 during M. tuberculosis infection is still unknown. Using IL-10 reporter mice, we show in this article that during the first 14 d of M. tuberculosis infection, the predominant cells expressing IL-10 in the lung were Ly6C+ monocytes. However, after day 21 postinfection, IL-10-expressing T cells were also highly represented. Notably, mice deficient in T cell-derived IL-10, but not mice deficient in monocyte-derived IL-10, showed a significant reduction in lung bacterial loads during chronic M. tuberculosis infection compared with fully IL-10-competent mice, indicating a major role for T cell-derived IL-10 in TB susceptibility. IL-10-expressing cells were detected among both CD4+ and CD8+ T cells, expressed high levels of CD44 and Tbet, and were able to coproduce IFN-γ and IL-10 upon ex vivo stimulation. Furthermore, during M. tuberculosis infection, Il10 expression in CD4+ T cells was partially regulated by both IL-27 and type I IFN signaling. Together, our data reveal that, despite the multiple immune sources of IL-10 during M. tuberculosis infection, activated effector T cells are the major source accounting for IL-10-induced TB susceptibility.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-10/immunology , Tuberculosis/immunology , Animals , Antigens, Ly/immunology , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukins/immunology , Interleukins/metabolism , Mice , Monocytes/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/microbiology
2.
PLoS Pathog ; 11(3): e1004715, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25763578

ABSTRACT

Interleukin-21 signaling is important for germinal center B-cell responses, isotype switching and generation of memory B cells. However, a role for IL-21 in antibody-mediated protection against pathogens has not been demonstrated. Here we show that IL-21 is produced by T follicular helper cells and co-expressed with IFN-γ during an erythrocytic-stage malaria infection of Plasmodium chabaudi in mice. Mice deficient either in IL-21 or the IL-21 receptor fail to resolve the chronic phase of P. chabaudi infection and P. yoelii infection resulting in sustained high parasitemias, and are not immune to re-infection. This is associated with abrogated P. chabaudi-specific IgG responses, including memory B cells. Mixed bone marrow chimeric mice, with T cells carrying a targeted disruption of the Il21 gene, or B cells with a targeted disruption of the Il21r gene, demonstrate that IL-21 from T cells signaling through the IL-21 receptor on B cells is necessary to control chronic P. chabaudi infection. Our data uncover a mechanism by which CD4+ T cells and B cells control parasitemia during chronic erythrocytic-stage malaria through a single gene, Il21, and demonstrate the importance of this cytokine in the control of pathogens by humoral immune responses. These data are highly pertinent for designing malaria vaccines requiring long-lasting protective B-cell responses.


Subject(s)
B-Lymphocytes/immunology , Immunity, Humoral/immunology , Interleukins/immunology , Malaria/immunology , Signal Transduction , T-Lymphocytes/immunology , Animals , Cell Communication/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasmodium chabaudi/immunology , Plasmodium yoelii/immunology , Real-Time Polymerase Chain Reaction
3.
Int J Parasitol ; 42(6): 549-55, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22549022

ABSTRACT

Despite intense research, malaria still is the one of the most devastating diseases killing more people than any other parasitic infection. In an attempt to control the infection, the host immune system produces a potent pro-inflammatory response. However, this response is also associated with complications, such as severe anaemia, hypoglycaemia and cerebral malaria. This pronounced production of pro-inflammatory cytokines response is a common feature of malaria caused by parasites infecting humans as well as rodents and primates. A balance between pro- and anti-inflammatory responses may be fundamental to the elimination of the parasite without inducing excessive host pathology. IL-10 is a key cytokine that has been shown to have an important regulatory function in establishing this balance in malaria. Here we discuss which cells can produce IL-10 during infection, and present an overview of the evidence showing that T-cell derived IL-10 plays an important role in regulating malaria pathology. Many different subsets of T cells can produce IL-10, however, evidence is accumulating that it is effector Th1 CD4(+) T cells which provide the crucial source that down-regulates inflammatory pathology during blood-stage malaria infections.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Host-Pathogen Interactions , Interleukin-10/immunology , Malaria/immunology , Malaria/pathology , Th1 Cells/immunology , Animals , Humans , Primates , Rodentia
4.
PLoS One ; 7(1): e29894, 2012.
Article in English | MEDLINE | ID: mdl-22272258

ABSTRACT

Plasmodium chabaudi infection induces a rapid and intense splenic CD4(+) T cell response that contributes to both disease pathogenesis and the control of acute parasitemia. The subsequent development of clinical immunity to disease occurs concomitantly with the persistence of low levels of chronic parasitemia. The suppressive activity of regulatory T (T(reg)) cells has been implicated in both development of clinical immunity and parasite persistence. To evaluate whether IL-2 is required to induce and to sustain the suppressive activity of T(reg) cells in malaria, we examined in detail the effects of anti-IL-2 treatment with JES6-1 monoclonal antibody (mAb) on the splenic CD4(+) T cell response during acute and chronic P. chabaudi AS infection in C57BL/6 mice. JES6-1 treatment on days 0, 2 and 4 of infection partially inhibits the expansion of the CD4(+)CD25(+)Foxp3(+) cell population during acute malaria. Despite the concomitant secretion of IL-2 and expression of high affinity IL-2 receptor by large CD4(+) T cells, JES6-1 treatment does not impair effector CD4(+) T cell activation and IFN-γ production. However, at the chronic phase of the disease, an enhancement of cellular and humoral responses occurs in JES6-1-treated mice, with increased production of TNF-α and parasite-specific IgG2a antibodies. Furthermore, JES6-1 mAb completely blocked the in vitro proliferation of CD4(+) T cells from non-treated chronic mice, while it further increased the response of CD4(+) T cells from JES6-1-treated chronic mice. We conclude that JES6-1 treatment impairs the expansion of T(reg) cell population during early P. chabaudi malaria and enhances the Th1 cell response in the late phase of the disease.


Subject(s)
Antibodies, Monoclonal/immunology , Interleukin-2/immunology , Malaria/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Chronic Disease , Female , Flow Cytometry , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Immunophenotyping , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2 Receptor beta Subunit/immunology , Interleukin-2 Receptor beta Subunit/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Malaria/drug therapy , Malaria/parasitology , Mice , Mice, Inbred C57BL , Parasitemia/drug therapy , Parasitemia/immunology , Parasitemia/parasitology , Plasmodium chabaudi/drug effects , Plasmodium chabaudi/immunology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism
5.
J Immunol ; 188(3): 1178-90, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22205023

ABSTRACT

Infection with the malaria parasite, Plasmodium, is characterized by excessive inflammation. The establishment of a precise balance between the pro- and anti-inflammatory responses is critical to guarantee control of the parasite and survival of the host. IL-10, a key regulatory cytokine produced by many cells of the immune system, has been shown to protect mice against pathology during acute Plasmodium0 chabaudi chabaudi AS model of malaria. However, the critical cellular source of IL-10 is still unknown. In this article, we demonstrate that T cell-derived IL-10 is necessary for the control of pathology during acute malaria, as mice bearing specific deletion of Il10 in T cells fully reproduce the phenotype observed in Il10(-)(/)(-) mice, with significant weight loss, decline in temperature, and increased mortality. Furthermore, we show that IFN-γ(+) Th1 cells are the main producers of IL-10 throughout acute infection, expressing high levels of CD44 and ICOS, and low levels of CD127. Although Foxp3(+) regulatory CD4(+) T cells produce IL-10 during infection, highly activated IFN-γ(+) Th1 cells were shown to be the essential and sufficient source of IL-10 to guarantee protection against severe immune-mediated pathology. Finally, in this model of malaria, we demonstrate that the generation of protective IL10(+)IFN-γ(+) Th1 cells is dependent on IL-27 signaling and independent of IL-21.


Subject(s)
Interleukin-10/biosynthesis , Interleukins/physiology , Malaria/immunology , Th1 Cells/metabolism , Animals , Inflammation , Interferon-gamma , Lymphocyte Activation/immunology , Malaria/pathology , Mice , Th1 Cells/immunology
6.
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
7.
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
8.
Blood ; 114(27): 5522-31, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-19837977

ABSTRACT

Host responses controlling blood-stage malaria include both innate and acquired immune effector mechanisms. During Plasmodium chabaudi infection in mice, a population of CD11b(high)Ly6C(+) monocytes are generated in bone marrow, most of which depend on the chemokine receptor CCR2 for migration from bone marrow to the spleen. In the absence of this receptor mice harbor higher parasitemias. Most importantly, splenic CD11b(high)Ly6C(+) cells from P chabaudi-infected wild-type mice significantly reduce acute-stage parasitemia in CCR2(-/-) mice. The CD11b(high)Ly6C(+) cells in this malaria infection display effector functions such as production of inducible nitric oxide synthase and reactive oxygen intermediates, and phagocytose P chabaudi parasites in vitro, and in a proportion of the cells, in vivo in the spleen, suggesting possible mechanisms of parasite killing. In contrast to monocyte-derived dendritic cells, CD11b(high)Ly6C(+) cells isolated from malaria-infected mice express low levels of major histocompatibility complex II and have limited ability to present the P chabaudi antigen, merozoite surface protein-1, to specific T-cell receptor transgenic CD4 T cells and fail to activate these T cells. We propose that these monocytes, which are rapidly produced in the bone marrow as part of the early defense mechanism against invading pathogens, are important for controlling blood-stage malaria parasites.


Subject(s)
Cell Movement/physiology , Monocytes/parasitology , Plasmodium chabaudi/physiology , Spleen/parasitology , Animals , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/parasitology , Antigen-Presenting Cells/pathology , Antigens, Ly/metabolism , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/parasitology , CD4-Positive T-Lymphocytes/pathology , Flow Cytometry , Host-Parasite Interactions , Malaria/blood , Malaria/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Monocytes/pathology , Nitric Oxide Synthase Type II/metabolism , Parasitemia/metabolism , Phagocytosis/physiology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Spleen/metabolism , Spleen/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/parasitology , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/metabolism
9.
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
...