Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
J Clin Invest ; 133(19)2023 10 02.
Article in English | MEDLINE | ID: mdl-37781920

ABSTRACT

The development of highly effective malaria vaccines and improvement of drug-treatment protocols to boost antiparasitic immunity are critical for malaria elimination. However, the rapid establishment of parasite-specific immune regulatory networks following exposure to malaria parasites hampers these efforts. Here, we identified stimulator of interferon genes (STING) as a critical mediator of type I interferon production by CD4+ T cells during blood-stage Plasmodium falciparum infection. The activation of STING in CD4+ T cells by cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) stimulated IFNB gene transcription, which promoted development of IL-10- and IFN-γ-coproducing CD4+ T (type I regulatory [Tr1]) cells. The critical role for type I IFN signaling for Tr1 cell development was confirmed in vivo using a preclinical malaria model. CD4+ T cell sensitivity to STING phosphorylation was increased in healthy volunteers following P. falciparum infection, particularly in Tr1 cells. These findings identified STING expressed by CD4+ T cells as an important mediator of type I IFN production and Tr1 cell development and activation during malaria.


Subject(s)
Interferon Type I , Malaria, Falciparum , T-Lymphocytes, Regulatory , Humans , CD4-Positive T-Lymphocytes , Interferon Type I/immunology , Malaria, Falciparum/immunology , T-Lymphocytes, Regulatory/immunology
4.
J Clin Invest ; 133(1)2023 01 03.
Article in English | MEDLINE | ID: mdl-36594463

ABSTRACT

Control of intracellular parasites responsible for malaria requires host IFN-γ+T-bet+CD4+ T cells (Th1 cells) with IL-10 produced by Th1 cells to mitigate the pathology induced by this inflammatory response. However, these IL-10-producing Th1 (induced type I regulatory [Tr1]) cells can also promote parasite persistence or impair immunity to reinfection or vaccination. Here, we identified molecular and phenotypic signatures that distinguished IL-10-Th1 cells from IL-10+Tr1 cells in Plasmodium falciparum-infected people who participated in controlled human malaria infection studies, as well as C57BL/6 mice with experimental malaria caused by P. berghei ANKA. We also identified a conserved Tr1 cell molecular signature shared between patients with malaria, dengue, and graft-versus-host disease. Genetic manipulation of primary human CD4+ T cells showed that the transcription factor cMAF played an important role in the induction of IL-10, while BLIMP-1 promoted the development of human CD4+ T cells expressing multiple coinhibitory receptors. We also describe heterogeneity of Tr1 cell coinhibitory receptor expression that has implications for targeting these molecules for clinical advantage during infection. Overall, this work provides insights into CD4+ T cell development during malaria that offer opportunities for creation of strategies to modulate CD4+ T cell functions and improve antiparasitic immunity.


Subject(s)
Malaria , T-Lymphocytes, Regulatory , Mice , Animals , Humans , Th1 Cells , Interleukin-10 , Mice, Inbred C57BL , Malaria/genetics , CD4-Positive T-Lymphocytes
5.
Nat Immunol ; 21(10): 1205-1218, 2020 10.
Article in English | MEDLINE | ID: mdl-32839608

ABSTRACT

Immune-modulating therapies have revolutionized the treatment of chronic diseases, particularly cancer. However, their success is restricted and there is a need to identify new therapeutic targets. Here, we show that natural killer cell granule protein 7 (NKG7) is a regulator of lymphocyte granule exocytosis and downstream inflammation in a broad range of diseases. NKG7 expressed by CD4+ and CD8+ T cells played key roles in promoting inflammation during visceral leishmaniasis and malaria-two important parasitic diseases. Additionally, NKG7 expressed by natural killer cells was critical for controlling cancer initiation, growth and metastasis. NKG7 function in natural killer and CD8+ T cells was linked with their ability to regulate the translocation of CD107a to the cell surface and kill cellular targets, while NKG7 also had a major impact on CD4+ T cell activation following infection. Thus, we report a novel therapeutic target expressed on a range of immune cells with functions in different immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Inflammation/immunology , Killer Cells, Natural/immunology , Leishmania donovani/physiology , Leishmaniasis, Visceral/immunology , Malaria/immunology , Membrane Proteins/metabolism , Plasmodium/physiology , Animals , Cells, Cultured , Cytotoxicity, Immunologic , Disease Models, Animal , Exocytosis , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Secretory Vesicles/metabolism
6.
Cell Rep ; 30(8): 2512-2525.e9, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32101732

ABSTRACT

Type I interferons (IFNs) play critical roles in anti-viral and anti-tumor immunity. However, they also suppress protective immune responses in some infectious diseases. Here, we identify type I IFNs as major upstream regulators of CD4+ T cells from visceral leishmaniasis (VL) patients. Furthermore, we report that mice deficient in type I IFN signaling have significantly improved control of Leishmania donovani, a causative agent of human VL, associated with enhanced IFNγ but reduced IL-10 production by parasite-specific CD4+ T cells. Importantly, we identify a small-molecule inhibitor that can be used to block type I IFN signaling during established infection and acts synergistically with conventional anti-parasitic drugs to improve parasite clearance and enhance anti-parasitic CD4+ T cell responses in mice and humans. Thus, manipulation of type I IFN signaling is a promising strategy for improving disease outcome in VL patients.


Subject(s)
Immunity/drug effects , Interferon Type I/pharmacology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Parasites/immunology , Amphotericin B/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Epitopes , Humans , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/pharmacology , Mice, Inbred C57BL , Nitriles , Parasites/drug effects , Pyrazoles/pharmacology , Pyrimidines , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/drug effects
7.
J Immunol ; 201(11): 3362-3372, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30355785

ABSTRACT

The outcome of intracellular parasitic infection can be determined by the immunoregulatory activities of natural regulatory CD4+ Foxp3+ T (Treg) cells and the anti-inflammatory cytokine IL-10. These mechanisms protect tissue but can also suppress antiparasitic CD4+ T cell responses. The specific contribution of these regulatory pathways during human parasitic diseases remains unclear. In this study, we investigated the roles of Treg cells and IL-10 during experimental visceral leishmaniasis caused by Leishmania donovani infection of C57BL/6 mice. We report only a limited contribution of Treg cells in suppressing antiparasitic immunity, but important roles in delaying the development of splenic pathology and restricting leukocyte expansion. We next employed a range of cell-specific, IL-10- and IL-10R-deficient mice and found these Treg cell functions were independent of IL-10. Instead, conventional CD4+ T cells and dendritic cells were the most important cellular sources of IL-10, and the absence of IL-10 in either cell population resulted in greater control of parasite growth but also caused accelerated breakdown in splenic microarchitecture. We also found that T cells, dendritic cells, and other myeloid cells were the main IL-10-responding cells because in the absence of IL-10R expression by these cell populations, there was greater expansion of parasite-specific CD4+ T cell responses associated with improved control of parasite growth. Again, however, there was also an accelerated breakdown in splenic microarchitecture in these animals. Together, these findings identify distinct, cell-specific, immunoregulatory networks established during experimental visceral leishmaniasis that could be manipulated for clinical advantage.


Subject(s)
Interleukin-10/metabolism , Leishmania donovani/physiology , Leishmaniasis, Visceral/immunology , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4 Antigens/metabolism , Cells, Cultured , Female , Forkhead Transcription Factors/metabolism , Humans , Immunomodulation , Mice , Mice, Inbred C57BL , Models, Animal
8.
Clin Transl Immunology ; 7(1): e1003, 2018.
Article in English | MEDLINE | ID: mdl-29484181

ABSTRACT

Objectives: Innate lymphoid cells (ILCs) share many characteristics with CD4+ T cells, and group 1 ILCs share a requirement for T-bet and the ability to produce IFNγ with T helper 1 (Th1) cells. Given this similarity, and the importance of Th1 cells for protection against intracellular protozoan parasites, we aimed to characterise the role of group 1 ILCs during Plasmodium infection. Methods: We quantified group 1 ILCs in peripheral blood collected from subjects infected with with Plasmodium falciparum 3D7 as part of a controlled human malaria infection study, and in the liver and spleens of Pc AS-infected mice. We used genetically-modified mouse models, as well as cell-depletion methods in mice to characterise the role of group 1 ILCs during Pc AS infection. Results: In a controlled human malaria infection study, we found that the frequencies of circulating ILC1s and NK cells decreased as infection progressed but recovered after volunteers were treated with antiparasitic drug. A similar observation was made for liver and splenic ILC1s in P. chabaudi chabaudi AS (Pc AS)-infected mice. The decrease in mouse liver ILC1 frequencies was associated with increased apoptosis. We also identified a population of cells within the liver and spleen that expressed both ILC1 and NK cell markers, indicative of plasticity between these two cell lineages. Studies using genetic and cell-depletion approaches indicated that group 1 ILCs have a limited role in antiparasitic immunity during Pc AS infection in mice. Discussion: Our results are consistent with a previous study indicating a limited role for natural killer (NK) cells during Plasmodium chabaudi infection in mice. Additionally, a recent study reported the redundancy of ILCs in humans with competent B and T cells. Nonetheless, our results do not rule out a role for group 1 ILCs in human malaria in endemic settings given that blood stage infection was initiated intravenously in our experimental models, and thus bypassed the liver stage of infection, which may influence the immune response during the blood stage. Conclusion: Our results show that ILC1s are lost early during mouse and human malaria, and this observation may help to explain the limited role for these cells in controlling blood stage infection.

9.
Front Immunol ; 8: 1307, 2017.
Article in English | MEDLINE | ID: mdl-29075269

ABSTRACT

Many infectious diseases are characterized by the development of immunoregulatory pathways that contribute to pathogen persistence and associated disease symptoms. In diseases caused by intracellular parasites, such as visceral leishmaniasis (VL), various immune modulators have the capacity to negatively impact protective CD4+ T cell functions. Galectin-1 is widely expressed on immune cells and has previously been shown to suppress inflammatory responses and promote the development of CD4+ T cells with immunoregulatory characteristics. Here, we investigated the role of galectin-1 in experimental VL caused by infection of C57BL/6 mice with Leishmania donovani. Mice lacking galectin-1 expression exhibited enhanced tissue-specific control of parasite growth in the liver, associated with an augmented Th1 cell response. However, unlike reports in other experimental models, we found little role for galectin-1 in the generation of IL-10-producing Th1 (Tr1) cells, and instead report that galectin-1 suppressed hepatic Th1 cell development. Furthermore, we found relatively early effects of galectin-1 deficiency on parasite growth, suggesting involvement of innate immune cells. However, experiments investigating the impact of galectin-1 deficiency on dendritic cells indicated that they were not responsible for the phenotypes observed in galectin-1-deficient mice. Instead, studies examining galectin-1 expression by CD4+ T cells supported a T cell intrinsic role for galectin-1 in the suppression of hepatic Th1 cell development during experimental VL. Together, our findings provide new information on the roles of galectin-1 during parasitic infection and indicate an important role for this molecule in tissue-specific Th1 cell development, but not CD4+ T cell IL-10 production.

10.
Sci Immunol ; 2(9)2017 Mar 03.
Article in English | MEDLINE | ID: mdl-28345074

ABSTRACT

Differentiation of naïve CD4+ T cells into functionally distinct T helper subsets is crucial for the orchestration of immune responses. Due to extensive heterogeneity and multiple overlapping transcriptional programs in differentiating T cell populations, this process has remained a challenge for systematic dissection in vivo. By using single-cell transcriptomics and computational analysis using a temporal mixtures of Gaussian processes model, termed GPfates, we reconstructed the developmental trajectories of Th1 and Tfh cells during blood-stage Plasmodium infection in mice. By tracking clonality using endogenous TCR sequences, we first demonstrated that Th1/Tfh bifurcation had occurred at both population and single-clone levels. Next, we identified genes whose expression was associated with Th1 or Tfh fates, and demonstrated a T-cell intrinsic role for Galectin-1 in supporting a Th1 differentiation. We also revealed the close molecular relationship between Th1 and IL-10-producing Tr1 cells in this infection. Th1 and Tfh fates emerged from a highly proliferative precursor that upregulated aerobic glycolysis and accelerated cell cycling as cytokine expression began. Dynamic gene expression of chemokine receptors around bifurcation predicted roles for cell-cell in driving Th1/Tfh fates. In particular, we found that precursor Th cells were coached towards a Th1 but not a Tfh fate by inflammatory monocytes. Thus, by integrating genomic and computational approaches, our study has provided two unique resources, a database www.PlasmoTH.org, which facilitates discovery of novel factors controlling Th1/Tfh fate commitment, and more generally, GPfates, a modelling framework for characterizing cell differentiation towards multiple fates.

11.
Cell Rep ; 17(2): 399-412, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27705789

ABSTRACT

The development of immunoregulatory networks is important to prevent disease. However, these same networks allow pathogens to persist and reduce vaccine efficacy. Here, we identify type I interferons (IFNs) as important regulators in developing anti-parasitic immunity in healthy volunteers infected for the first time with Plasmodium falciparum. Type I IFNs suppressed innate immune cell function and parasitic-specific CD4+ T cell IFNγ production, and they promoted the development of parasitic-specific IL-10-producing Th1 (Tr1) cells. Type I IFN-dependent, parasite-specific IL-10 production was also observed in P. falciparum malaria patients in the field following chemoprophylaxis. Parasite-induced IL-10 suppressed inflammatory cytokine production, and IL-10 levels after drug treatment were positively associated with parasite burdens before anti-parasitic drug administration. These findings have important implications for understanding the development of host immune responses following blood-stage P. falciparum infection, and they identify type I IFNs and related signaling pathways as potential targets for therapies or vaccine efficacy improvement.


Subject(s)
Host-Parasite Interactions/immunology , Immunity, Innate/genetics , Interferon Type I/genetics , Malaria, Falciparum/immunology , Antiparasitic Agents/administration & dosage , CD4-Positive T-Lymphocytes/immunology , Healthy Volunteers , Humans , Interferon Type I/immunology , Interferon-gamma/genetics , Interleukin-10/genetics , Interleukin-10/immunology , Malaria, Falciparum/drug therapy , Malaria, Falciparum/genetics , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Th1 Cells/immunology , Th1 Cells/metabolism
13.
PLoS Negl Trop Dis ; 10(2): e0004415, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26872334

ABSTRACT

Chronic disease caused by infections, cancer or autoimmunity can result in profound immune suppression. Immunoregulatory networks are established to prevent tissue damage caused by inflammation. Although these immune checkpoints preserve tissue function, they allow pathogens and tumors to persist, and even expand. Immune checkpoint blockade has recently been successfully employed to treat cancer. This strategy modulates immunoregulatory mechanisms to allow host immune cells to kill or control tumors. However, the utility of this approach for controlling established infections has not been extensively investigated. Here, we examined the potential of modulating glucocorticoid-induced TNF receptor-related protein (GITR) on T cells to improve anti-parasitic immunity in blood and spleen tissue from visceral leishmaniasis (VL) patients infected with Leishmania donovani. We found little effect on parasite growth or parasite-specific IFNγ production. However, this treatment reversed the improved anti-parasitic immunity achieved by IL-10 signaling blockade. Further investigations using an experimental VL model caused by infection of C57BL/6 mice with L. donovani revealed that this negative effect was prominent in the liver, dependent on parasite burden and associated with an accumulation of Th1 cells expressing high levels of KLRG-1. Nevertheless, combined anti-IL-10 and anti-GITR mAb treatment could improve anti-parasitic immunity when used with sub-optimal doses of anti-parasitic drug. However, additional studies with VL patient samples indicated that targeting GITR had no overall benefit over IL-10 signaling blockade alone at improving anti-parasitic immune responses, even with drug treatment cover. These findings identify several important factors that influence the effectiveness of immune modulation, including parasite burden, target tissue and the use of anti-parasitic drug. Critically, these results also highlight potential negative effects of combining different immune modulation strategies.


Subject(s)
Immunotherapy , Leishmania donovani/physiology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/therapy , Animals , Cytokines/immunology , Female , Humans , Interleukin-10/immunology , Leishmaniasis, Visceral/parasitology , Mice , Mice, Inbred C57BL , Spleen/immunology , Spleen/parasitology , Th1 Cells/immunology
14.
PLoS Pathog ; 12(1): e1005398, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26765224

ABSTRACT

Tumor necrosis factor (TNF) is critical for controlling many intracellular infections, but can also contribute to inflammation. It can promote the destruction of important cell populations and trigger dramatic tissue remodeling following establishment of chronic disease. Therefore, a better understanding of TNF regulation is needed to allow pathogen control without causing or exacerbating disease. IL-10 is an important regulatory cytokine with broad activities, including the suppression of inflammation. IL-10 is produced by different immune cells; however, its regulation and function appears to be cell-specific and context-dependent. Recently, IL-10 produced by Th1 (Tr1) cells was shown to protect host tissues from inflammation induced following infection. Here, we identify a novel pathway of TNF regulation by IL-10 from Tr1 cells during parasitic infection. We report elevated Blimp-1 mRNA levels in CD4+ T cells from visceral leishmaniasis (VL) patients, and demonstrate IL-12 was essential for Blimp-1 expression and Tr1 cell development in experimental VL. Critically, we show Blimp-1-dependent IL-10 production by Tr1 cells prevents tissue damage caused by IFNγ-dependent TNF production. Therefore, we identify Blimp-1-dependent IL-10 produced by Tr1 cells as a key regulator of TNF-mediated pathology and identify Tr1 cells as potential therapeutic tools to control inflammation.


Subject(s)
Inflammation/immunology , Interleukin-10/biosynthesis , Leishmaniasis, Visceral/immunology , Repressor Proteins/immunology , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Disease Models, Animal , Female , Flow Cytometry , Humans , Inflammation/pathology , Interleukin-10/immunology , Leishmaniasis, Visceral/pathology , Malaria/immunology , Malaria/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Fluorescence , Positive Regulatory Domain I-Binding Factor 1 , T-Lymphocytes, Regulatory/immunology
15.
J Immunol ; 195(12): 5707-17, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26538396

ABSTRACT

Intracellular infections, such as those caused by the protozoan parasite Leishmania donovani, a causative agent of visceral leishmaniasis (VL), require a potent host proinflammatory response for control. IL-17 has emerged as an important proinflammatory cytokine required for limiting growth of both extracellular and intracellular pathogens. However, there are conflicting reports on the exact roles for IL-17 during parasitic infections and limited knowledge about cellular sources and the immune pathways it modulates. We examined the role of IL-17 in an experimental model of VL caused by infection of C57BL/6 mice with L. donovani and identified an early suppressive role for IL-17 in the liver that limited control of parasite growth. IL-17-producing γδ T cells recruited to the liver in the first week of infection were the critical source of IL-17 in this model, and CCR2(+) inflammatory monocytes were an important target for the suppressive effects of IL-17. Improved parasite control was independent of NO generation, but associated with maintenance of superoxide dismutase mRNA expression in the absence of IL-17 in the liver. Thus, we have identified a novel inhibitory function for IL-17 in parasitic infection, and our results demonstrate important interactions among γδ T cells, monocytes, and infected macrophages in the liver that can determine the outcome of parasitic infection.


Subject(s)
Interleukin-17/metabolism , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Liver/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Humans , Immunosuppression Therapy , Leishmania donovani/growth & development , Liver/parasitology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/parasitology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, CCR2/metabolism , Superoxide Dismutase/metabolism , T-Lymphocytes/parasitology
16.
Front Immunol ; 5: 498, 2014.
Article in English | MEDLINE | ID: mdl-25352846

ABSTRACT

CD4(+) T cells are critical for defense against protozoan parasites. Intracellular protozoan parasite infections generally require the development of a Th1 cell response, characterized by the production of IFNγ and TNF that are critical for the generation of microbicidal molecules by phagocytes, as well as the expression of cytokines and cell surface molecules needed to generate cytolytic CD8(+) T cells that can recognize and kill infected host cells. Over the past 25 years, much has been learnt about the molecular and cellular components necessary for the generation of Th1 cell responses, and it has become clear that these responses need to be tightly controlled to prevent disease. However, our understanding of the immunoregulatory mechanisms activated during infection is still not complete. Furthermore, it is apparent that although these mechanisms are critical to prevent inflammation, they can also promote parasite persistence and development of disease. Here, we review how CD4(+) T cells are controlled during protozoan infections and how these regulatory mechanisms can influence parasite growth and disease outcome.

17.
J Clin Invest ; 124(6): 2483-96, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24789914

ABSTRACT

Many pathogens, including viruses, bacteria, and protozoan parasites, suppress cellular immune responses through activation of type I IFN signaling. Recent evidence suggests that immune suppression and susceptibility to the malaria parasite, Plasmodium, is mediated by type I IFN; however, it is unclear how type I IFN suppresses immunity to blood-stage Plasmodium parasites. During experimental severe malaria, CD4+ Th cell responses are suppressed, and conventional DC (cDC) function is curtailed through unknown mechanisms. Here, we tested the hypothesis that type I IFN signaling directly impairs cDC function during Plasmodium infection in mice. Using cDC-specific IFNAR1-deficient mice, and mixed BM chimeras, we found that type I IFN signaling directly affects cDC function, limiting the ability of cDCs to prime IFN-γ-producing Th1 cells. Although type I IFN signaling modulated all subsets of splenic cDCs, CD8- cDCs were especially susceptible, exhibiting reduced phagocytic and Th1-promoting properties in response to type I IFNs. Additionally, rapid and systemic IFN-α production in response to Plasmodium infection required type I IFN signaling in cDCs themselves, revealing their contribution to a feed-forward cytokine-signaling loop. Together, these data suggest abrogation of type I IFN signaling in CD8- splenic cDCs as an approach for enhancing Th1 responses against Plasmodium and other type I IFN-inducing pathogens.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/metabolism , Malaria/immunology , Th1 Cells/immunology , Animals , CD8 Antigens/metabolism , Dendritic Cells/classification , Female , Immune Tolerance , Immunity, Cellular , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Plasmodium berghei/immunology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/immunology
18.
J Immunol ; 192(8): 3709-18, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24634490

ABSTRACT

Organ-specific immunity is a feature of many infectious diseases, including visceral leishmaniasis caused by Leishmania donovani. Experimental visceral leishmaniasis in genetically susceptible mice is characterized by an acute, resolving infection in the liver and chronic infection in the spleen. CD4+ T cell responses are critical for the establishment and maintenance of hepatic immunity in this disease model, but their role in chronically infected spleens remains unclear. In this study, we show that dendritic cells are critical for CD4+ T cell activation and expansion in all tissue sites examined. We found that FTY720-mediated blockade of T cell trafficking early in infection prevented Ag-specific CD4+ T cells from appearing in lymph nodes, but not the spleen and liver, suggesting that early CD4+ T cell priming does not occur in liver-draining lymph nodes. Extended treatment with FTY720 over the first month of infection increased parasite burdens, although this associated with blockade of lymphocyte egress from secondary lymphoid tissue, as well as with more generalized splenic lymphopenia. Importantly, we demonstrate that CD4+ T cells are required for the establishment and maintenance of antiparasitic immunity in the liver, as well as for immune surveillance and suppression of parasite outgrowth in chronically infected spleens. Finally, although early CD4+ T cell priming appeared to occur most effectively in the spleen, we unexpectedly revealed that protective CD4+ T cell-mediated hepatic immunity could be generated in the complete absence of all secondary lymphoid tissues.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Animals , Antigens, Protozoan/immunology , CD4-Positive T-Lymphocytes/drug effects , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , Female , Fingolimod Hydrochloride , Immunosuppressive Agents/pharmacology , Liver/drug effects , Liver/immunology , Liver/parasitology , Lymphocyte Activation/immunology , Lymphoid Tissue/drug effects , Lymphoid Tissue/immunology , Lymphoid Tissue/parasitology , Mice , Mice, Knockout , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Spleen/drug effects , Spleen/immunology , Spleen/parasitology
19.
PLoS Pathog ; 7(10): e1002279, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21998581

ABSTRACT

LIGHT (TNFSF14) is a member of the TNF superfamily involved in inflammation and defence against infection. LIGHT signals via two cell-bound receptors; herpes virus entry mediator (HVEM) and lymphotoxin-beta receptor (LTßR). We found that LIGHT is critical for control of hepatic parasite growth in mice with visceral leishmaniasis (VL) caused by infection with the protozoan parasite Leishmania donovani. LIGHT-HVEM signalling is essential for early dendritic cell IL-12/IL-23p40 production, and the generation of IFNγ- and TNF-producing T cells that control hepatic infection. However, we also discovered that LIGHT-LTßR interactions suppress anti-parasitic immunity in the liver in the first 7 days of infection by mechanisms that restrict both CD4(+) T cell function and TNF-dependent microbicidal mechanisms. Thus, we have identified distinct roles for LIGHT in infection, and show that manipulation of interactions between LIGHT and its receptors may be used for therapeutic advantage.


Subject(s)
Immunity, Cellular , Leishmania donovani/pathogenicity , Leishmaniasis, Visceral/pathology , Receptors, Tumor Necrosis Factor, Member 14/metabolism , T-Lymphocytes/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Cell Proliferation/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-12/biosynthesis , Interleukin-23/biosynthesis , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Liver/parasitology , Liver/pathology , Lymphotoxin beta Receptor/immunology , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Member 14/immunology , Signal Transduction , T-Lymphocytes/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...