Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Int Immunol ; 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38895753

ABSTRACT

The generation and maintenance of memory T cells are regulated by various factors, including cytokines. Previous studies have shown that IL-27 is produced during the early acute phase of Plasmodium chabaudi chabaudi AS (Pcc) infection and inhibits the development of Th1-type memory CD4+ T cells. However, whether IL-27 acts directly on its receptor on Plasmodium-specific CD4+ T cells or indirectly via its receptor on other immune cells remains unclear. We aimed to determine the role of IL-27 receptor signaling in different immune cell types in regulating the generation and phenotype of memory CD4+ T cells during Plasmodium infection. We utilized Plasmodium-specific TCR transgenic mice, PbT-II, and Il27rα-/- mice to assess the direct and indirect effects of IL-27 signaling on memory CD4+ T-cell generation. Mice were transferred with PbT-II or Il27rα-/- PbT-II cells and infected with Pcc. Conditional knockout mice lacking the IL-27 receptor in T cells or dendritic cells were employed to discern the specific immune cell types involved in IL-27 receptor signaling. High levels of memory in PbT-II cells with Th1-shift occurred only when both PbT-II and host cells lacked the IL-27 receptor, suggesting the predominant inhibitory role of IL-27 signaling in both cell types. Furthermore, IL-27 receptor signaling in T cells limited the number of memory CD4+ T cells, while signaling in both T and dendritic cells contributed to the Th1 dominance of memory CD4+ T cells. These findings underscore the complex cytokine signaling network regulating memory CD4+ T cells during Plasmodium infection.

2.
Immunity ; 44(3): 672-682, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26968425

ABSTRACT

Interleukin-27 (IL-27) is a heterodimeric regulatory cytokine of the IL-12 family, which is produced by macrophages, dendritic cells, and B cells upon stimulation through innate immune receptors. Here, we described regulatory CD4(+) T cells that produce IL-27 in response to T cell receptor stimulation during malaria infection, inhibiting IL-2 production and clonal expansion of other T cells in an IL-27-dependent manner. IL-27-producing CD4(+) T cells were Foxp3(-)CD11a(+)CD49d(+) malaria antigen-specific CD4(+) T cells and were distinct from interferon-γ (IFN-γ) producing Th1 or IL-10 producing Tr1 cells. In mice lacking IL-27 in T cells, IL-2 production was restored and clonal expansion and IFN-γ production by specific CD4(+) T cells were improved, culminating in reduced parasite burden. This study highlights a unique population of IL-27 producing regulatory CD4(+) T cells and their critical role in the regulation of the protective immune response against malaria parasites.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Interleukin-27/metabolism , Malaria/immunology , Plasmodium berghei/immunology , T-Lymphocytes, Regulatory/physiology , Animals , CD4-Positive T-Lymphocytes/parasitology , Cell Proliferation/genetics , Cells, Cultured , Cytokines/metabolism , Immunity, Innate , Interleukin-27/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, Interleukin , T-Lymphocytes, Regulatory/parasitology
3.
Int Immunol ; 34(1): 21-33, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34648636

ABSTRACT

Plasmodium parasites that infect humans are highly polymorphic, and induce various infections ranging from an asymptomatic state to life-threatening diseases. However, how the differences between the parasites affect host immune responses during blood-stage infection remains largely unknown. We investigated the CD4+ T-cell immune responses in mice infected with P. berghei ANKA (PbA) or P. chabaudi chabaudi AS (Pcc) using PbT-II cells, which recognize a common epitope of these parasites. In the acute phase of infection, CD4+ T-cell responses in PbA-infected mice showed a lower involvement of Th1 cells and a lower proportion of Ly6Clo effector CD4+ T cells than those in Pcc-infected mice. Transcriptome analysis of PbT-II cells indicated that type I interferon (IFN)-regulated genes were expressed at higher levels in both Th1- and Tfh-type PbT-II cells from PbA-infected mice than those from Pcc-infected mice. Moreover, IFN-α levels were considerably higher in PbA-infected mice than in Pcc-infected mice. Inhibition of type I IFN signaling increased PbT-II and partially reversed the Th1 over Tfh bias of the PbT-II cells in both PbA- and Pcc-infected mice. In the memory phase, PbT-II cells in PbA-primed mice maintained higher numbers and exhibited a better recall response to the antigen. However, recall responses were not significantly different between the infection groups after re-challenge with PbA, suggesting the effect of the inflammatory environment by the infection. These observations suggest that the differences in Plasmodium-specific CD4+ T-cell responses between PbA- and Pcc-infected mice were associated with the difference in type I IFN production during the early phase of the infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interferon Type I/biosynthesis , Malaria/immunology , Plasmodium berghei/immunology , Plasmodium chabaudi/immunology , Animals , Cells, Cultured , Mice , Mice, Transgenic
4.
Int Immunol ; 33(8): 409-422, 2021 07 23.
Article in English | MEDLINE | ID: mdl-33914894

ABSTRACT

Upon activation, specific CD4+ T cells up-regulate the expression of CD11a and CD49d, surrogate markers of pathogen-specific CD4+ T cells. However, using T-cell receptor transgenic mice specific for a Plasmodium antigen, termed PbT-II, we found that activated CD4+ T cells develop not only to CD11ahiCD49dhi cells, but also to CD11ahiCD49dlo cells during acute Plasmodium infection. CD49dhi PbT-II cells, localized in the red pulp of spleens, expressed transcription factor T-bet and produced IFN-γ, indicating that they were type 1 helper T (Th1)-type cells. In contrast, CD49dlo PbT-II cells resided in the white pulp/marginal zones and were a heterogeneous population, with approximately half of them expressing CXCR5 and a third expressing Bcl-6, a master regulator of follicular helper T (Tfh) cells. In adoptive transfer experiments, both CD49dhi and CD49dlo PbT-II cells differentiated into CD49dhi Th1-type cells after stimulation with antigen-pulsed dendritic cells, while CD49dhi and CD49dlo phenotypes were generally maintained in mice infected with Plasmodium chabaudi. These results suggest that CD49d is expressed on Th1-type Plasmodium-specific CD4+ T cells, which are localized in the red pulp of the spleen, and can be used as a marker of antigen-specific Th1 CD4+ T cells, rather than that of all pathogen-specific CD4+ T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Integrin alpha4/immunology , Malaria/immunology , Plasmodium chabaudi/immunology , T Follicular Helper Cells/immunology , Th1 Cells/immunology , Adoptive Transfer/methods , Animals , Cells, Cultured , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-6/immunology , Spleen/immunology , T-Lymphocytes, Helper-Inducer/immunology
5.
Eur J Immunol ; 48(8): 1319-1328, 2018 08.
Article in English | MEDLINE | ID: mdl-29745988

ABSTRACT

Interferon regulatory factor 4 (IRF4) has critical roles in immune cell differentiation and function and is indispensable for clonal expansion and effector function in T cells. Here, we demonstrate that the AKT pathway is impaired in murine CD8+ T cells lacking IRF4. The expression of phosphatase and tensin homolog (PTEN), a negative regulator of the AKT pathway, was elevated in Irf4-/- CD8+ T cells. Inhibition of PTEN partially rescued downstream events, suggesting that PTEN constitutes a checkpoint in the IRF4-mediated regulation of cell signaling. Despite the clonal expansion defect, in the absence of IRF4, memory-like CD8+ T cells could be generated and maintained, although unable to expand in recall responses. The homeostatic proliferation of naïve Irf4-/- CD8+ T cells was impaired, whereas their number eventually reached a level similar to that of wild-type CD8+ T cells. Conversely, memory-like Irf4-/- CD8+ T cells underwent homeostatic proliferation in a manner similar to that of wild-type memory CD8+ T cells. These results suggest that IRF4 regulates the clonal expansion of CD8+ T cells at least in part via the AKT signaling pathway. Moreover, IRF4 regulates the homeostatic proliferation of naïve CD8+ T cells, whereas the maintenance of memory CD8+ T cells is IRF4-independent.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interferon Regulatory Factors/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/immunology , Animals , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Interferon Regulatory Factors/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , PTEN Phosphohydrolase/antagonists & inhibitors , Signal Transduction/immunology
6.
Genes Cells ; 23(10): 828-838, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30112853

ABSTRACT

Histone H2A phosphorylation plays a role both in chromatin condensation during mitosis and in transcriptional activation during the G1/S transition. Bub1 and NHK1/VRK1 have been identified as histone H2A kinases. However, little is known about the importance of histone H2A phosphorylation in chromosome segregation. Here, we expressed recombinant hBUB1 and confirmed that it phosphorylates histone H2A T120 in the in vitro-assembled nucleosome. Knockdown (KD) of BUB1 decreases bulk H2A T120 phosphorylation in HeLa cells, whereas hBUB1 is upregulated during mitosis, which corresponds with H2A T120 phosphorylation. ChIP-qPCR of the DXZ1 centromeric and γ-ALR pericentromeric region showed that BUB1 localizes to this region and increases local H2A T120 phosphorylation during M phase. BUB1 KD did not induce apoptosis but increased the M phase cell population, as detected by flow cytometry. BUB1 KD also caused an abnormal metaphase and telophase, resulting in multinucleated cells and impaired cancer cell growth both in vitro and in vivo. Over-expression of the histone H2A T120D or T120E mutations, which mimic phosphorylated threonine, decreased the number of multinucleated cells caused by BUB1 KD. These results strengthen the apparent importance of BUB1-mediated H2A T120 phosphorylation in normal mitosis.


Subject(s)
Chromosome Segregation/physiology , Histones/genetics , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/metabolism , Centromere/metabolism , Centromere/physiology , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation/genetics , Gene Knockdown Techniques/methods , HeLa Cells , Heterochromatin , Histones/metabolism , Humans , Interphase , Kinetochores/metabolism , Mitosis , Phosphorylation , Threonine
7.
Infect Immun ; 86(4)2018 04.
Article in English | MEDLINE | ID: mdl-29426043

ABSTRACT

CD8+ T cells are the major effector cells that protect against malaria liver-stage infection, forming clusters around Plasmodium-infected hepatocytes and eliminating parasites after a prolonged interaction with these hepatocytes. We aimed to investigate the roles of specific and nonspecific CD8+ T cells in cluster formation and protective immunity. To this end, we used Plasmodium berghei ANKA expressing ovalbumin as well as CD8+ T cells from transgenic mice expressing a T cell receptor specific for ovalbumin (OT-I) and CD8+ T cells specific for an unrelated antigen, respectively. While antigen-specific CD8+ T cells were essential for cluster formation, both antigen-specific and nonspecific CD8+ T cells joined the clusters. However, nonspecific CD8+ T cells did not significantly contribute to protective immunity. In the livers of infected mice, specific CD8+ T cells expressed high levels of CD25, compatible with a local, activated effector phenotype. In vivo imaging of the liver revealed that specific CD8+ T cells interact with CD11c+ cells around infected hepatocytes. The depletion of CD11c+ cells virtually eliminated the clusters in the liver, leading to a significant decrease in protection. These experiments reveal an essential role of hepatic CD11c+ dendritic cells and presumably macrophages in the formation of CD8+ T cell clusters around Plasmodium-infected hepatocytes. Once cluster formation is triggered by parasite-specific CD8+ T cells, specific and unrelated activated CD8+ T cells join the clusters in a chemokine- and dendritic cell-dependent manner. Nonspecific CD8+ T cells seem to play a limited role in protective immunity against Plasmodium parasites.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Liver Diseases, Parasitic/immunology , Macrophages/immunology , Malaria/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Hepatocytes/immunology , Liver Diseases, Parasitic/diagnosis , Liver Diseases, Parasitic/parasitology , Lymphocyte Activation/immunology , Macrophages/metabolism , Malaria/diagnosis , Malaria/parasitology , Mice , Mice, Transgenic
8.
Microbiol Immunol ; 60(2): 121-31, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26696540

ABSTRACT

CD4(+) T cells play critical roles in protection against the blood stage of malarial infection; however, their uncontrolled activation can be harmful to the host. In this study, in which rodent models of Plasmodium parasites were used, the expression of inhibitory receptors on activated CD4(+) T cells and their cytokine production was compared with their expression in a bacterial and another protozoan infection. CD4(+) T cells from mice infected with P. yoelii 17XL, P yoelii 17XNL, P. chabaudi, P. vinckei and P. berghei expressed the inhibitory receptors, PD-1 and LAG-3, as early as 6 days after infection, whereas those from either Listeria monocytogenes- or Leishmania major-infected mice did not. In response to T-cell receptor stimulation, CD4(+) T cells from mice infected with all the pathogens under study produced high concentrations of IFN-γ. IL-2 production was reduced in mice infected with Plasmodium species, but not in those infected with Listeria or Leishmania. In vitro blockade of the interaction between PD-1 and its ligands resulted in increased IFN-γ production in response to Plasmodium antigens, implying that PD-1 expressed on activated CD4(+) T cells actively inhibits T cell immune responses. Studies using Myd88(-/-), Trif(-/-) and Irf3(-/-) mice showed that induction of these CD4(+) T cells and their ability to produce cytokines is largely independent of TLR signaling. These studies suggest that expression of the inhibitory receptors PD-1 and LAG-3 on CD4(+) T cells and their reduced IL-2 production are common characteristic features of Plasmodium infection.


Subject(s)
Antigens, CD/biosynthesis , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Malaria/immunology , Plasmodium/immunology , Programmed Cell Death 1 Receptor/biosynthesis , Animals , Antigen-Presenting Cells/immunology , Antigens, CD/immunology , Cytokines/immunology , Interleukin-2/biosynthesis , Interleukin-2/immunology , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Listeria monocytogenes/immunology , Listeriosis/immunology , Lymphocyte Activation , Malaria/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/immunology , Lymphocyte Activation Gene 3 Protein
9.
J Immunol ; 192(5): 2271-9, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24489086

ABSTRACT

IRF4 is a transcription factor from the IRF factor family that plays pivotal roles in the differentiation and function of T and B lymphocytes. Although IRF4 is also expressed in dendritic cells (DCs) and macrophages, its roles in these cells in vivo are not clearly understood. In this study, conditional knockout mice that lack IRF4 in DCs or macrophages were generated and infected with Leishmania major. Mice lacking DC expression of IRF4 showed reduced footpad swelling compared with C57BL/6 mice, whereas those lacking IRF4 in macrophages did not. Mice with IRF4-deficient DCs also showed reduced parasite burden, and their CD4(+) T cells produced higher levels of IFN-γ in response to L. major Ag. In the draining lymph nodes, the proportion of activated CD4(+) T cells in these mice was similar to that in the control, but the proportion of IFN-γ-producing cells was increased, suggesting a Th1 bias in the immune response. Moreover, the numbers of migrating Langerhans cells and other migratory DCs in the draining lymph nodes were reduced both before and postinfection in mice with IRF4 defects in DCs, but higher levels of IL-12 were observed in IRF4-deficient DCs. These results imply that IRF4 expression in DCs inhibits their ability to produce IL-12 while promoting their migratory behavior, thus regulating CD4(+) T cell responses against local infection with L. major.


Subject(s)
Interferon Regulatory Factors/immunology , Interleukin-12/immunology , Langerhans Cells/immunology , Leishmania major/metabolism , Leishmaniasis, Cutaneous/immunology , Th1 Cells/immunology , Animals , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Interferon Regulatory Factors/genetics , Interferon-gamma/immunology , Interleukin-12/genetics , Langerhans Cells/pathology , Leishmaniasis, Cutaneous/genetics , Leishmaniasis, Cutaneous/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Th1 Cells/pathology
10.
J Immunol ; 189(9): 4396-404, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23008449

ABSTRACT

Conditions required for establishing protective immune memory vary depending on the infecting microbe. Although the memory immune response against malaria infection is generally thought to be relatively slow to develop and can be lost rapidly, experimental evidence is insufficient. In this report, we investigated the generation, maintenance, and recall responses of Ag-specific memory CD8(+) T cells using Plasmodium berghei ANKA expressing OVA (PbA-OVA) as a model system. Mice were transferred with OVA-specific CD8(+) T (OT-I) cells and infected with PbA-OVA or control Listeria monocytogenes expressing OVA (LM-OVA). Central memory type OT-I cells were maintained for >2 mo postinfection and recovery from PbA-OVA. Memory OT-I cells produced IFN-γ as well as TNF-α upon activation and were protective against challenge with a tumor expressing OVA, indicating that functional memory CD8(+) T cells can be generated and maintained postinfection with P. berghei ANKA. Cotransfer of memory OT-I cells with naive OT-I cells to mice followed by infection with PbA-OVA or LM-OVA revealed that clonal expansion of memory OT-I cells was limited during PbA-OVA infection compared with expansion of naive OT-I cells, whereas it was more rapid during LM-OVA infection. The expression of inhibitory receptors programmed cell death-1 and LAG-3 was higher in memory-derived OT-I cells than naive-derived OT-I cells during infection with PbA-OVA. These results suggest that memory CD8(+) T cells can be established postinfection with P. berghei ANKA, but their recall responses during reinfection are more profoundly inhibited than responses of naive CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/parasitology , Immunologic Memory , Malaria/immunology , Plasmodium berghei/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Cell Line, Tumor , Epitopes, T-Lymphocyte/metabolism , Female , Listeria monocytogenes/immunology , Malaria/blood , Malaria/parasitology , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
11.
Infect Immun ; 81(10): 3825-34, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23897612

ABSTRACT

Following Anopheles mosquito-mediated introduction into a human host, Plasmodium parasites infect hepatocytes and undergo intensive replication. Accumulating evidence indicates that CD8(+) T cells induced by immunization with attenuated Plasmodium sporozoites can confer sterile immunity at the liver stage of infection; however, the mechanisms underlying this protection are not clearly understood. To address this, we generated recombinant Plasmodium berghei ANKA expressing a fusion protein of an ovalbumin epitope and green fluorescent protein in the cytoplasm of the parasite. We have shown that the ovalbumin epitope is presented by infected liver cells in a manner dependent on a transporter associated with antigen processing and becomes a target of specific CD8(+) T cells from the T cell receptor transgenic mouse line OT-I, leading to protection at the liver stage of Plasmodium infection. We visualized the interaction between OT-I cells and infected hepatocytes by intravital imaging using two-photon microscopy. OT-I cells formed clusters around infected hepatocytes, leading to the elimination of the intrahepatic parasites and subsequent formation of large clusters of OT-I cells in the liver. Gamma interferon expressed in CD8(+) T cells was dispensable for this protective response. Additionally, we found that polyclonal ovalbumin-specific memory CD8(+) T cells induced by de novo immunization were able to confer sterile protection, although the threshold frequency of the protection was relatively high. These studies revealed a novel mechanism of specific CD8(+) T cell-mediated protective immunity and demonstrated that proteins expressed in the cytoplasm of Plasmodium parasites can become targets of specific CD8(+) T cells during liver-stage infection.


Subject(s)
Antigens, Protozoan/physiology , CD8-Positive T-Lymphocytes/physiology , Hepatocytes/parasitology , Plasmodium berghei/metabolism , Animals , Gene Expression Regulation , Humans , Liver , Malaria , Mice , Mice, Transgenic , Nucleoproteins , Plasmodium berghei/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
12.
Microbiol Immunol ; 57(3): 213-23, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23278848

ABSTRACT

The spleen is the main organ for immune defense during infection with Plasmodium parasites and splenomegaly is one of the major symptoms of such infections. Using a rodent model of Plasmodium yoelii infection, MHC class II(+)CD11c(-) non-T, non-B cells in the spleen were characterized. Although the proportion of conventional dendritic cells was reduced, that of MHC II(+)CD11c(-) non-T, non-B cells increased during the course of infection. The increase in this subpopulation was dependent on the presence of lymphocytes. Experiments using Rag-2(-/-) mice with adoptively transferred normal spleen cells indicated that these cells were non-lymphoid cells; however, their accumulation in the spleen during infection with P. yoelii depended on lymphocytes. Functionally, these MHC II(+)CD11c(-) non-T, non-B cells were able to produce the proinflammatory cytokines alpha tumor necrosis factor and interleukin-6 in response to infected red blood cells, but had only a limited ability to activate antigen-specific CD4(+) T cells. This study revealed a novel interaction between MHC II(+)CD11c(-) non-lymphoid cells and lymphoid cells in the accumulations of these non-lymphoid cells in the spleen during infection with P. yoelii.


Subject(s)
CD11c Antigen/analysis , Histocompatibility Antigens Class II/analysis , Leukocytes/immunology , Malaria/immunology , Plasmodium yoelii/immunology , Spleen/immunology , Animals , Disease Models, Animal , Interleukin-6/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism
13.
Biol Pharm Bull ; 36(11): 1800-6, 2013.
Article in English | MEDLINE | ID: mdl-24189423

ABSTRACT

In this experiment, we developed a novel safe and effective gene delivery vector coated with γ-polyglutamic acid (γ-PGA-coated complexes). The γ-PGA-coated complex was composed of chiseled spherical nano-particles with anionic charges. The plasmid DNA/polyethyleneimine complex (non-coated complex) showed high transgene efficiency in the spleen and lung after intravenous administration in mice, with high liver toxicity and lethality. On the other hand, γ-PGA-coated complex selectively showed high transgene efficiency in the spleen without such toxicity. Furthermore, the γ-PGA-coated complex highly accumulated and showed high gene expression in the marginal zone of the spleen. Those results strongly indicated that γ-PGA-coated complex was suitable as a DNA vaccine vector. We therefore applied γ-PGA-coated complex to melanoma DNA vaccine, pUb-M. The γ-PGA-coated complex containing pUb-M significantly inhibited the growth and metastasis of a melanoma cell line, B16-F10 cells. In conclusion, we developed a splenic gene vector, γ-PGA-coated complex, as a novel technology for clinical vaccination.


Subject(s)
Cancer Vaccines , Gene Transfer Techniques , Polyglutamic Acid/analogs & derivatives , Spleen/metabolism , Vaccines, DNA , Animals , Cell Line, Tumor , DNA/administration & dosage , DNA/chemistry , Liver/drug effects , Liver/pathology , Luciferases/genetics , Luciferases/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Plasmids , Polyethyleneimine/administration & dosage , Polyethyleneimine/chemistry , Polyglutamic Acid/administration & dosage , Polyglutamic Acid/chemistry , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology
14.
Int Immunol ; 22(12): 941-52, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21059770

ABSTRACT

T-cell immune responses are critical for protection of the host and for disease pathogenesis during infection with Plasmodium species. We examined the regulation of CD4(+) T-cell cytokine responses during infection with Plasmodium berghei ANKA (PbA). CD4(+) T cells from PbA-infected mice produced IFN-γ, IL-4 and IL-10 in response to TCR stimulation at levels higher than those from uninfected mice. This altered cytokine response was dependent on parasitemia. To examine the specificity of the response, mice were adoptively transferred with CD4(+) T cells from OT-II TCR transgenic mice and were infected with PbA expressing OVA. Unexpectedly, CD4(+) T cells from the OT-II-transferred wild-type PbA-infected mice showed high levels of IFN-γ production after stimulation with OVA and the cells producing IFN-γ were not OT-II but were host CD4(+) T cells. Further investigation revealed that host CD4(+) T cells produced IFN-γ in response to IL-2 produced by activated OT-II cells. This IFN-γ response was completely inhibited by anti-CD25 mAbs, and this effect was not due to the block of the survival signals provided by IL-2. Furthermore, IFN-γ production by CD4(+) T cells in response to PbA antigens was dependent on IL-2. These findings suggest the importance of IL-2 levels during infection with malaria parasites and indicate that CD4(+) T cells can produce IFN-γ without TCR engagement via a bystander mechanism in response to IL-2 produced by other activated CD4(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interferon-gamma/biosynthesis , Interleukin-2/immunology , Malaria/immunology , Plasmodium berghei/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/biosynthesis , Lymphocyte Count , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/immunology
15.
Proc Natl Acad Sci U S A ; 105(41): 15890-5, 2008 Oct 14.
Article in English | MEDLINE | ID: mdl-18836070

ABSTRACT

Interferon regulatory factor (IRF) 4 is a member of the IRF family of transcription factors and plays critical roles in the development of CD4(+) T cells into Th2 and Th17 cells. Using the infection model of Nippostrongyrus brasiliensis, we have confirmed the critical roles of IRF-4 in Th2 development in vivo by using IRF-4(-/-) BALB/c mice. However, naïve IRF-4(-/-)CD4(+) T cells produced Th2 cytokines, including IL-4, IL-5, and IL-10, but not IL-2 or IFN-gamma, at levels higher than wild-type BALB/c CD4(+) T cells in response to T cell receptor stimulation. In contrast, effector/memory IRF-4(-/-)CD4(+) T cells did not exhibit increased production of Th2 cytokines. Knockdown of IRF-4 expression by using small interfering RNA promoted IL-4 production in naïve CD4(+) T cells but inhibited it in effector/memory CD4(+) T cells. These results indicate that IRF-4 plays differential roles in the regulation of Th2 cytokine production in naïve CD4(+) T cells and effector/memory CD4(+) T cells. IRF-4 inhibits Th2 cytokine production in naïve CD4(+) T cells, whereas it promotes Th2 cytokine production in effector/memory CD4(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Gene Expression Regulation/immunology , Interferon Regulatory Factors/physiology , Animals , Immunologic Memory , Interferon Regulatory Factors/deficiency , Mice , Mice, Inbred BALB C , Mice, Knockout , Th2 Cells/immunology
16.
J Immunol ; 181(2): 1420-8, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606696

ABSTRACT

Cerebral malaria is one of the severe complications of Plasmodium falciparum infection. Studies using a rodent model of Plasmodium berghei ANKA infection established that CD8(+) T cells are involved in the pathogenesis of cerebral malaria. However, it is unclear whether and how Plasmodium-specific CD8(+) T cells can be activated during the erythrocyte stage of malaria infection. We generated recombinant Plasmodium berghei ANKA expressing OVA (OVA-PbA) to investigate the parasite-specific T cell responses during malaria infection. Using this model system, we demonstrate two types of CD8(+) T cell activations during the infection with malaria parasite. Ag (OVA)-specific CD8(+) T cells were activated by TAP-dependent cross-presentation during infection with OVA-PbA leading to their expression of an activation phenotype and granzyme B and the development to functional CTL. These highly activated CD8(+) T cells were preferentially sequestered in the brain, although it was unclear whether these cells were involved in the pathogenesis of cerebral malaria. Activation of OVA-specific CD8(+) T cells in RAG2 knockout TCR-transgenic mice during infection with OVA-PbA did not have a protective role but rather was pathogenic to the host as shown by their higher parasitemia and earlier death when compared with RAG2 knockout mice. The OVA-specific CD8(+) T cells, however, were also activated during infection with wild-type parasites in an Ag-nonspecific manner, although the levels of activation were much lower. This nonspecific activation occurred in a TAP-independent manner, appeared to require NK cells, and was not by itself pathogenic to the host.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , Malaria, Cerebral/immunology , Malaria/immunology , Plasmodium berghei/immunology , Animals , Cross-Priming , Interferon-gamma/blood , Interferon-gamma/immunology , Interferon-gamma/metabolism , Malaria, Cerebral/blood , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Parasitemia , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology
17.
Parasitol Int ; 74: 101994, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31634628

ABSTRACT

IL-27, a regulatory cytokine, plays critical roles in the prevention of immunopathology during Plasmodium infection. We examined these roles in the immune responses against Plasmodium chabaudi infection using the Il-27ra-/- mice. While IL-27 was expressed at high levels during the early phase of the infection, enhanced CD4+ T cell function and reduction in parasitemia were observed mainly during the chronic phase in the mutant mice. In mice infected with P. chabaudi and cured with drug, CD4+ T cells in the Il-27ra-/- mice exhibited enhanced CD4+ T-cell responses, indicating the inhibitory role of IL-27 on the protective immune responses. To determine the role of IL-27 in detail, we performed CD4+ T-cell transfer experiments. The Il-27ra-/- and Il27p28-/- mice were first infected with P. chabaudi and then cured using drug treatment. Plasmodium-antigen primed CD4+ T cells were prepared from these mice and transferred into the recipient mice, followed by infection with the heterologous parasite P. berghei ANKA. Il-27ra-/- CD4+ T cells in the infected recipient mice did not produce IL-10, indicating that IL-10 production by primed CD4+ T cells is IL-27 dependent. Il27p28-/- CD4+ T cells that were primed in the absence of IL-27 exhibited enhanced recall responses during the challenge infection with P. berghei ANKA, implying that IL-27 receptor signaling during the primary infection affects recall responses in the long-term via the regulation of the memory CD4+ T cell generation. These features highlighted direct and time-transcending roles of IL-27 in the regulation of immune responses against chronic infection with Plasmodium parasites.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Immunologic Memory , Interleukin-10/immunology , Interleukin-27/genetics , Malaria/immunology , Animals , Chronic Disease , Interleukin-27/immunology , Malaria/drug therapy , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Plasmodium chabaudi
18.
Parasitol Int ; 70: 5-15, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30639137

ABSTRACT

Malaria is induced by infection with Plasmodium parasites, which are genetically diverse, and the immune response to Plasmodium infection has both allele-specific and cross-reactive components. To determine the role of the cross-reactive immune response in the protection and disease manifestation in heterologous Plasmodium infection, we used infection models of P. chabaudi chabaudi (Pcc) and P. berghei ANKA (PbA). CD4+ T cells primed with Pcc infection exhibited strong cross-reactivity to PbA antigens. We infected C57BL/6 mice with Pcc and subsequently treated them with an anti-Plasmodium drug. The Pcc-primed mice exhibited reduced parasitemia and showed no signs of experimental cerebral malaria after infection with PbA. CD4+ T cells from the Pcc-primed mice produced high levels of IFN-γ and IL-10 in response to PbA early after PbA infection. The blockade of IL-10 signaling with anti-IL-10 receptor antibody increased the proportion of activated CD4+ and γδ T cells and the IFN-γ production by CD4+ T cells in response to PbA antigens, while markedly reducing the levels of parasitemia. In contrast, IL-10 blockade did not have a significant effect on parasitemia levels in unprimed mice after PbA infection. These data suggest a potent regulatory role of IL-10 in the cross-reactive memory response to the infection with heterologous Plasmodium parasites leading to the inhibition of the protective immunity and pathogenesis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cross Reactions , Interleukin-10/immunology , Malaria, Cerebral/immunology , Animals , Gene Expression Regulation , Immunologic Memory , Interferon-gamma/immunology , Interleukin-10/genetics , Malaria, Cerebral/drug therapy , Mice , Mice, Inbred C57BL , Parasitemia/immunology , Plasmodium/pathogenicity , Plasmodium berghei
19.
Front Immunol ; 9: 2942, 2018.
Article in English | MEDLINE | ID: mdl-30619302

ABSTRACT

Adaptive immune responses are critical for protection against infection with Plasmodium parasites. The metabolic state dramatically changes in T cells during activation and the memory phase. Recent findings suggest that metformin, a medication for treating type-II diabetes, enhances T-cell immune responses by modulating lymphocyte metabolism. In this study, we investigated whether metformin could enhance anti-malaria immunity. Mice were infected with Plasmodium yoelii and administered metformin. Levels of parasitemia were reduced in treated mice compared with those in untreated mice, starting at ~2 weeks post-infection. The number of γδ T cells dramatically increased in the spleens of treated mice compared with that in untreated mice during the later phase of infection, while that of αß T cells did not. The proportions of Vγ1+ and Vγ2+ γδ T cells increased, suggesting that activated cells were selectively expanded. However, these γδ T cells expressed inhibitory receptors and had severe defects in cytokine production, suggesting that they were in a state of exhaustion. Metformin was unable to rescue the cells from exhaustion at this stage. Depletion of γδ T cells with antibody treatment did not affect the reduction of parasitemia in metformin-treated mice, suggesting that the effect of metformin on the reduction of parasitemia was independent of γδ T cells.


Subject(s)
Malaria/drug therapy , Metformin/pharmacology , Parasitemia/drug therapy , Plasmodium yoelii/immunology , T-Lymphocyte Subsets/drug effects , Animals , Disease Models, Animal , Female , Humans , Lymphocyte Activation/drug effects , Malaria/immunology , Malaria/parasitology , Metformin/therapeutic use , Mice , Mice, Inbred C57BL , Parasitemia/immunology , Parasitemia/parasitology , Plasmodium yoelii/pathogenicity , Receptors, Antigen, T-Cell, gamma-delta/antagonists & inhibitors , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome
20.
Parasitol Int ; 66(3): 227-235, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28163249

ABSTRACT

The spleen is the major organ in which T cells are primed during infection with malaria parasites. However, little is known regarding the dynamics of the immune responses and their localization within the splenic tissue during malaria infection. We examined murine CD8+ T cell responses during infection with Plasmodium berghei using recombinant parasites expressing a model antigen ovalbumin (OVA) protein and compared the responses with those elicited by Listeria monocytogenes expressing the same antigen. OVA-specific CD8+ T cells were mainly activated in the white pulp of the spleen during malaria infection, as similarly observed during Listeria infection. However, the fates of these activated CD8+ T cells were distinct. During infection with malaria parasites, activated CD8+ T cells preferentially accumulated in the red pulp and/or marginal zone, where cytokine production of OVA-specific CD8+ T cells decreased, and the expression of multiple inhibitory receptors increased. These cells preferentially underwent apoptosis, suggesting that T cell exhaustion mainly occurred in the red pulp and/or marginal zone. However, during Listeria infection, OVA-specific CD8+ T cells only transiently expressed inhibitory receptors in the white pulp and maintained their ability to produce cytokines and become memory cells. These results highlighted the distinct fates of CD8+ T cells during infection with Plasmodium parasites and Listeria, and suggested that activation and exhaustion of specific CD8+ T cells occurred in distinct spleen compartments during infection with malaria parasites.


Subject(s)
Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Malaria/immunology , Plasmodium berghei/immunology , Spleen/immunology , Animals , Antigens, Protozoan/genetics , Apoptosis , CD8-Positive T-Lymphocytes/physiology , Immunologic Memory , Interferon-gamma/immunology , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Lymphocyte Activation , Malaria/parasitology , Mice , Mice, Inbred C57BL , Ovalbumin/genetics , Ovalbumin/immunology , Plasmodium berghei/genetics , Spleen/cytology
SELECTION OF CITATIONS
SEARCH DETAIL