Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
J Immunol ; 207(7): 1836-1847, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34479944

ABSTRACT

DEC-205 is a cell-surface receptor that transports bound ligands into the endocytic pathway for degradation or release within lysosomal endosomes. This receptor has been reported to bind a number of ligands, including keratin, and some classes of CpG oligodeoxynucleotides (ODN). In this study, we explore in detail the requirements for binding ODNs, revealing that DEC-205 efficiently binds single-stranded, phosphorothioated ODN of ≥14 bases, with preference for the DNA base thymidine, but with no requirement for a CpG motif. DEC-205 fails to bind double-stranded phosphodiester ODN, and thus does not bind the natural type of DNA found in mammals. The ODN binding preferences of DEC-205 result in strong binding of B class ODN, moderate binding to C class ODN, minimal binding to P class ODN, and no binding to A class ODN. Consistent with DEC-205 binding capacity, induction of serum IL-12p70 or activation of B cells by each class of ODN correlated with DEC-205 dependence in mice. Thus, the greater the DEC-205 binding capacity, the greater the dependence on DEC-205 for optimal responses. Finally, by covalently linking a B class ODN that efficiently binds DEC-205, to a P class ODN that shows poor binding, we improved DEC-205 binding and increased adjuvancy of the hybrid ODN. The hybrid ODN efficiently enhanced induction of effector CD8 T cells in a DEC-205-dependent manner. Furthermore, the hybrid ODN induced robust memory responses, and was particularly effective at promoting the development of liver tissue-resident memory T cells.


Subject(s)
Adjuvants, Immunologic , Oligodeoxyribonucleotides , Animals , Dendritic Cells , Interleukin-12 , Liver , Mice
2.
J Exp Med ; 218(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33914023

ABSTRACT

Tissue-resident memory T cells (TRM cells) are key elements of tissue immunity. Here, we investigated the role of the regulator of T cell receptor and cytokine signaling, Ptpn2, in the formation and function of TRM cells in skin. Ptpn2-deficient CD8+ T cells displayed a marked defect in generating CD69+ CD103+ TRM cells in response to herpes simplex virus type 1 (HSV-1) skin infection. This was accompanied by a reduction in the proportion of KLRG1- memory precursor cells and a transcriptional bias toward terminal differentiation. Of note, forced expression of KLRG1 was sufficient to impede TRM cell formation. Normalizing memory precursor frequencies by transferring equal numbers of KLRG1- cells restored TRM generation, demonstrating that Ptpn2 impacted skin seeding with precursors rather than downstream TRM cell differentiation. Importantly, Ptpn2-deficient TRM cells augmented skin autoimmunity but also afforded superior protection from HSV-1 infection. Our results emphasize that KLRG1 repression is required for optimal TRM cell formation in skin and reveal an important role of Ptpn2 in regulating TRM cell functionality.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Lectins, C-Type/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/immunology , Receptors, Immunologic/immunology , Animals , Autoimmunity/immunology , Female , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Mice , Mice, Inbred C57BL , Skin/immunology
3.
J Immunol ; 205(7): 1842-1856, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32839238

ABSTRACT

Follicular dendritic cells and macrophages have been strongly implicated in presentation of native Ag to B cells. This property has also occasionally been attributed to conventional dendritic cells (cDC) but is generally masked by their essential role in T cell priming. cDC can be divided into two main subsets, cDC1 and cDC2, with recent evidence suggesting that cDC2 are primarily responsible for initiating B cell and T follicular helper responses. This conclusion is, however, at odds with evidence that targeting Ag to Clec9A (DNGR1), expressed by cDC1, induces strong humoral responses. In this study, we reveal that murine cDC1 interact extensively with B cells at the border of B cell follicles and, when Ag is targeted to Clec9A, can display native Ag for B cell activation. This leads to efficient induction of humoral immunity. Our findings indicate that surface display of native Ag on cDC with access to both T and B cells is key to efficient humoral vaccination.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Lectins, C-Type/metabolism , Receptors, Immunologic/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Antigen Presentation , Autoantigens/immunology , Autoantigens/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Immunity, Humoral , Lectins, C-Type/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Immunologic/genetics , Vaccination
5.
Cell Rep ; 25(1): 68-79.e4, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30282039

ABSTRACT

Liver tissue-resident memory T (Trm) cells migrate throughout the sinusoids and are capable of protecting against malaria sporozoite challenge. To gain an understanding of liver Trm cell development, we examined various conditions for their formation. Although liver Trm cells were found in naive mice, their presence was dictated by antigen specificity and required IL-15. Liver Trm cells also formed after adoptive transfer of in vitro-activated but not naive CD8+ T cells, indicating that activation was essential but that antigen presentation within the liver was not obligatory. These Trm cells patrolled the liver sinusoids with a half-life of 36 days and occupied a large niche that could be added to sequentially without effect on subsequent Trm cell cohorts. Together, our findings indicate that liver Trm cells form as a normal consequence of CD8+ T cell activation during essentially any infection but that inflammatory and antigenic signals preferentially tailor their development.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Liver/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/cytology , Epitopes , Hepatitis/immunology , Interleukin-15/immunology , Liver/cytology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL
6.
J Immunol ; 200(4): 1443-1456, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29321276

ABSTRACT

Differentiation of CD4+ Th cells is critical for immunity to malaria. Several innate immune signaling pathways have been implicated in the detection of blood-stage Plasmodium parasites, yet their influence over Th cell immunity remains unclear. In this study, we used Plasmodium-reactive TCR transgenic CD4+ T cells, termed PbTII cells, during nonlethal P. chabaudi chabaudi AS and P. yoelii 17XNL infection in mice, to examine Th cell development in vivo. We found no role for caspase1/11, stimulator of IFN genes, or mitochondrial antiviral-signaling protein, and only modest roles for MyD88 and TRIF-dependent signaling in controlling PbTII cell expansion. In contrast, IFN regulatory factor 3 (IRF3) was important for supporting PbTII expansion, promoting Th1 over T follicular helper (Tfh) differentiation, and controlling parasites during the first week of infection. IRF3 was not required for early priming by conventional dendritic cells, but was essential for promoting CXCL9 and MHC class II expression by inflammatory monocytes that supported PbTII responses in the spleen. Thereafter, IRF3-deficiency boosted Tfh responses, germinal center B cell and memory B cell development, parasite-specific Ab production, and resolution of infection. We also noted a B cell-intrinsic role for IRF3 in regulating humoral immune responses. Thus, we revealed roles for IRF3 in balancing Th1- and Tfh-dependent immunity during nonlethal infection with blood-stage Plasmodium parasites.


Subject(s)
Cell Differentiation/immunology , Interferon Regulatory Factor-3/immunology , Malaria/immunology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Animals , Female , Germinal Center/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/immunology
7.
J Immunol ; 199(12): 4165-4179, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29084838

ABSTRACT

We describe an MHC class II (I-Ab)-restricted TCR transgenic mouse line that produces CD4+ T cells specific for Plasmodium species. This line, termed PbT-II, was derived from a CD4+ T cell hybridoma generated to blood-stage Plasmodium berghei ANKA (PbA). PbT-II cells responded to all Plasmodium species and stages tested so far, including rodent (PbA, P. berghei NK65, Plasmodium chabaudi AS, and Plasmodium yoelii 17XNL) and human (Plasmodium falciparum) blood-stage parasites as well as irradiated PbA sporozoites. PbT-II cells can provide help for generation of Ab to P. chabaudi infection and can control this otherwise lethal infection in CD40L-deficient mice. PbT-II cells can also provide help for development of CD8+ T cell-mediated experimental cerebral malaria (ECM) during PbA infection. Using PbT-II CD4+ T cells and the previously described PbT-I CD8+ T cells, we determined the dendritic cell (DC) subsets responsible for immunity to PbA blood-stage infection. CD8+ DC (a subset of XCR1+ DC) were the major APC responsible for activation of both T cell subsets, although other DC also contributed to CD4+ T cell responses. Depletion of CD8+ DC at the beginning of infection prevented ECM development and impaired both Th1 and follicular Th cell responses; in contrast, late depletion did not affect ECM. This study describes a novel and versatile tool for examining CD4+ T cell immunity during malaria and provides evidence that CD4+ T cell help, acting via CD40L signaling, can promote immunity or pathology to blood-stage malaria largely through Ag presentation by CD8+ DC.


Subject(s)
Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , Dendritic Cells/immunology , Malaria/immunology , Mice, Transgenic/immunology , Parasitemia/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Protozoan/immunology , CD40 Antigens/deficiency , CD40 Ligand/immunology , Cells, Cultured , Crosses, Genetic , Hybridomas , Lymphocyte Activation , Malaria, Cerebral/immunology , Malaria, Cerebral/prevention & control , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic/genetics , Plasmodium berghei/immunology , Radiation Chimera
8.
Immunity ; 45(4): 889-902, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27692609

ABSTRACT

In recent years, various intervention strategies have reduced malaria morbidity and mortality, but further improvements probably depend upon development of a broadly protective vaccine. To better understand immune requirement for protection, we examined liver-stage immunity after vaccination with irradiated sporozoites, an effective though logistically difficult vaccine. We identified a population of memory CD8+ T cells that expressed the gene signature of tissue-resident memory T (Trm) cells and remained permanently within the liver, where they patrolled the sinusoids. Exploring the requirements for liver Trm cell induction, we showed that by combining dendritic cell-targeted priming with liver inflammation and antigen recognition on hepatocytes, high frequencies of Trm cells could be induced and these cells were essential for protection against malaria sporozoite challenge. Our study highlights the immune potential of liver Trm cells and provides approaches for their selective transfer, expansion, or depletion, which may be harnessed to control liver infections or autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Liver/immunology , Malaria/immunology , Animals , CD8-Positive T-Lymphocytes/parasitology , Culicidae , Dendritic Cells/immunology , Dendritic Cells/parasitology , Hepatocytes/immunology , Hepatocytes/parasitology , Liver/parasitology , Liver Diseases/immunology , Liver Diseases/parasitology , Malaria Vaccines/immunology , Mice , Plasmodium berghei/immunology , Sporozoites/immunology , Sporozoites/parasitology , Vaccination/methods
9.
Cell Rep ; 14(3): 586-597, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26774484

ABSTRACT

DCs often require stimulation from CD4(+) T cells to propagate CD8(+) T cell responses, but precisely how T cell help optimizes the priming capacity of DCs and why this appears to differ between varying types of CD8(+) T cell immunity remains unclear. We show that CD8(+) T cell priming upon HSV-1 skin infection depended on DCs receiving stimulation from both IFN-α/ß and CD4(+) T cells to provide IL-15. This was not an additive effect but resulted from CD4(+) T cells amplifying DC production of IL-15 in response to IFN-α/ß. We also observed that increased innate stimulation reversed the helper dependence of CD8(+) T cell priming and that the innate stimulus, rather than the CD4(+) T cells themselves, determined how "help'" was integrated into the priming response by DCs. These findings identify T cell help as a flexible means to amplify varying suboptimal innate signals in DCs.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Chemokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Herpesvirus 1, Human/physiology , Humans , Interferon-alpha/genetics , Interferon-alpha/metabolism , Interferon-alpha/pharmacology , Interferon-beta/metabolism , Interleukin-15/metabolism , Interleukin-6/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Skin Diseases/pathology , Skin Diseases/virology , T-Lymphocytes, Helper-Inducer/immunology
10.
J Immunol ; 195(3): 1006-14, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26101322

ABSTRACT

Targeting Ags to dendritic cell (DC) surface receptors can induce a variety of responses depending on the DC type targeted, the receptor targeted, and the adjuvant used. Clec9A (DNGR-1), which is expressed by CD8(+) DCs, has been shown to bind F-actin exposed on damaged cells. Targeting Ag to this receptor in mice and nonhuman primates induces strong humoral immunity even in the absence of adjuvant, a process seen for a few select DC receptors. In contrast with other receptors, however, targeting Clec9A induces long-lived, affinity-matured Ab responses that are associated with efficient CD4(+) T cell responses shown to possess properties of follicular Th cells (TFH). In this article, we provide definitive evidence that Clec9A targeting promotes the development of TFH by showing that responding CD4 T cells express CXCR5, PD1, the TFH transcription factor Bcl6, and the cytokine IL-21, and that these cells localize to germinal centers. Furthermore, we extend studies from the model Ag OVA to the viral Ag glycoprotein D of HSV-1 and examine the capacity of primed TFH to form functional memory. We show that targeting glycoprotein D to Clec9A even in the absence of adjuvant induced long-lived memory CXCR5(+) PD1(hi) CD4(+) T cells that proliferated extensively upon secondary challenge and rapidly developed into effector TFH. This was associated with enhanced germinal center B cell responses and accelerated Ab production. Our study indicates that targeting Ags to Clec9A in the absence of adjuvant routinely generates TFH responses that form long-lived memory capable of robust secondary TFH responses.


Subject(s)
Dendritic Cells/immunology , Immunologic Memory/immunology , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , Receptors, Immunologic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer , Animals , Antigens/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , DNA-Binding Proteins/biosynthesis , Germinal Center/cytology , Germinal Center/immunology , Interleukin-21 Receptor alpha Subunit/genetics , Interleukins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Programmed Cell Death 1 Receptor/biosynthesis , Proto-Oncogene Proteins c-bcl-6 , Receptors, CXCR5/biosynthesis , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/transplantation , Viral Envelope Proteins/immunology
11.
PLoS Pathog ; 10(5): e1004135, 2014 May.
Article in English | MEDLINE | ID: mdl-24854165

ABSTRACT

To follow the fate of CD8+ T cells responsive to Plasmodium berghei ANKA (PbA) infection, we generated an MHC I-restricted TCR transgenic mouse line against this pathogen. T cells from this line, termed PbT-I T cells, were able to respond to blood-stage infection by PbA and two other rodent malaria species, P. yoelii XNL and P. chabaudi AS. These PbT-I T cells were also able to respond to sporozoites and to protect mice from liver-stage infection. Examination of the requirements for priming after intravenous administration of irradiated sporozoites, an effective vaccination approach, showed that the spleen rather than the liver was the main site of priming and that responses depended on CD8α+ dendritic cells. Importantly, sequential exposure to irradiated sporozoites followed two days later by blood-stage infection led to augmented PbT-I T cell expansion. These findings indicate that PbT-I T cells are a highly versatile tool for studying multiple stages and species of rodent malaria and suggest that cross-stage reactive CD8+ T cells may be utilized in liver-stage vaccine design to enable boosting by blood-stage infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunization, Secondary/methods , Life Cycle Stages/immunology , Malaria/prevention & control , Plasmodium berghei/immunology , Receptors, Antigen, T-Cell/genetics , Sporozoites/immunology , Adoptive Transfer , Animals , Anopheles , Blood/parasitology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Liver/immunology , Liver/parasitology , Malaria/blood , Malaria/immunology , Malaria/parasitology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasmodium berghei/growth & development , Plasmodium chabaudi , Plasmodium yoelii , Receptors, Antigen, T-Cell/immunology
12.
Mol Immunol ; 53(4): 450-2, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23142929

ABSTRACT

Fluorescent proteins can be used to visualize cells and their constituents by various imaging techniques. Adoptive transfer of T cells from C57Bl/6 (B6) mice that expressed DsRed.T3 under the actin promoter lead to frequent rejection of transferred cells. In short term in vivo cytotoxicity assays these mice showed detectable, but weak lysis of DsRed spleen cells but their responses could be boosted by re-challenge with DsRed spleen cells. To determine whether DsRed protein may contain an H-2(b) MHC I-restricted T cell epitope, B6 mice immune to DsRed spleen cells were examined for in vivo lysis of target cells coated with various DsRed-derived peptides selected by the SYFPEITHI epitope prediction program. This analysis identified one D(b)-restricted peptide sequence within DsRed (SSLQDGCFI) that acted as an epitope for B6 target lysis. Knowledge of this epitope could allow DsRed to be used as a model antigen in B6 mice and cautions against using this fluorochrome, as well as several others containing the immunogenic sequence, in adoptive transfer studies where rejection is not desirable.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Luminescent Proteins/immunology , Peptides/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Amino Acid Sequence , Animals , Cytotoxicity Tests, Immunologic , Epitopes, T-Lymphocyte/chemistry , Fluorescent Dyes/chemistry , Gene Expression , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Luminescent Proteins/chemistry , Mice , Mice, Inbred C57BL , Molecular Mimicry , Molecular Sequence Data , Peptides/chemistry , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation
13.
J Immunol ; 189(2): 701-10, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22685317

ABSTRACT

Tight regulation of virus-induced cytotoxic effector CD8(+) T cells is essential to prevent immunopathology. Naturally occurring effector CD8(+) T cells, with a KLRG1(hi) CD62L(lo) phenotype typical of short-lived effector CD8(+) T cells (SLECs), can be found in increased numbers in autoimmune-prone mice, most notably in mice homozygous for the san allele of Roquin. These SLEC-like cells were able to trigger autoimmune diabetes in a susceptible background. When Roquin is mutated (Roquin(san)), effector CD8(+) T cells accumulate in a cell-autonomous manner, most prominently as SLEC-like effectors. Excessive IFN-γ promotes the accumulation of SLEC-like cells, increases their T-bet expression, and enhances their granzyme B production in vivo. We show that overexpression of IFN-γ was caused by failed posttranscriptional repression of Ifng mRNA. This study identifies a novel mechanism that prevents accumulation of self-reactive cytotoxic effectors, highlighting the importance of regulating Ifng mRNA stability to maintain CD8(+) T cell homeostasis and prevent CD8-mediated autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Aggregation/immunology , Cytotoxicity, Immunologic , Down-Regulation/immunology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/genetics , RNA, Messenger/antagonists & inhibitors , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , Cell Aggregation/genetics , Cellular Senescence/genetics , Cellular Senescence/immunology , Cytotoxicity, Immunologic/genetics , Down-Regulation/genetics , Homeostasis/genetics , Homeostasis/immunology , Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/metabolism , Interferon-gamma/biosynthesis , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation/immunology , RNA Stability/immunology , RNA, Messenger/genetics , Receptors, Immunologic , Trans-Activators/biosynthesis , Trans-Activators/genetics , Ubiquitin-Protein Ligases/genetics
14.
J Biol Chem ; 287(24): 20652-63, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22535952

ABSTRACT

Eukaryotic cells generate energy in the form of ATP, through a network of mitochondrial complexes and electron carriers known as the oxidative phosphorylation system. In mammals, mitochondrial complex I (CI) is the largest component of this system, comprising 45 different subunits encoded by mitochondrial and nuclear DNA. Humans diagnosed with mutations in the gene NDUFS4, encoding a nuclear DNA-encoded subunit of CI (NADH dehydrogenase ubiquinone Fe-S protein 4), typically suffer from Leigh syndrome, a neurodegenerative disease with onset in infancy or early childhood. Mitochondria from NDUFS4 patients usually lack detectable NDUFS4 protein and show a CI stability/assembly defect. Here, we describe a recessive mouse phenotype caused by the insertion of a transposable element into Ndufs4, identified by a novel combined linkage and expression analysis. Designated Ndufs4(fky), the mutation leads to aberrant transcript splicing and absence of NDUFS4 protein in all tissues tested of homozygous mice. Physical and behavioral symptoms displayed by Ndufs4(fky/fky) mice include temporary fur loss, growth retardation, unsteady gait, and abnormal body posture when suspended by the tail. Analysis of CI in Ndufs4(fky/fky) mice using blue native PAGE revealed the presence of a faster migrating crippled complex. This crippled CI was shown to lack subunits of the "N assembly module", which contains the NADH binding site, but contained two assembly factors not present in intact CI. Metabolomic analysis of the blood by tandem mass spectrometry showed increased hydroxyacylcarnitine species, implying that the CI defect leads to an imbalanced NADH/NAD(+) ratio that inhibits mitochondrial fatty acid ß-oxidation.


Subject(s)
DNA Transposable Elements , Electron Transport Complex I/metabolism , Leigh Disease/enzymology , Mitochondria/enzymology , Mutation , NAD/metabolism , Animals , Binding Sites , Electron Transport Complex I/genetics , Humans , Leigh Disease/genetics , Leigh Disease/pathology , Leigh Disease/physiopathology , Metabolomics/methods , Mice , Mice, Mutant Strains , Mice, Transgenic , Mitochondria/genetics , Mitochondria/pathology , NAD/genetics , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Proteomics/methods , RNA Splicing/genetics
15.
J Infect Dis ; 204(12): 1989-96, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21998471

ABSTRACT

Murine cerebral malaria is a complex disease caused by Plasmodium berghei ANKA infection. Several cell types, including CD8(+) T cells, are essential effectors of disease. Although the use of transgenic parasites expressing model antigens has revealed the induction of cytotoxic T lymphocytes (CTL) specific for these model antigens, there is no direct evidence for a response to authentic blood-stage parasite antigens, nor any knowledge of its magnitude. Our studies show that there is a dramatic primary parasite-specific CTL response, akin to viral immunity, reaching approximately 30% of splenic CD8(+) T cells, with many producing interferon-γ and tumor necrosis factor-α. These cells express granzyme B and other markers of specific responders, are cytolytic, and respond to a broad array of major histocompatibility complex (MHC) I-restricted epitopes, 5 of which are identified here. Our studies indicate that vigorous CTL responses can be induced to pathogens even when they largely reside in red blood cells, which lack MHC I processing machinery.


Subject(s)
Dendritic Cells/immunology , Plasmodium berghei/immunology , Schizonts/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , CD11a Antigen/metabolism , CD8 Antigens/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/parasitology , Epitope Mapping , Epitopes/immunology , Granzymes/metabolism , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Plasmodium berghei/growth & development , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/parasitology , Tumor Necrosis Factor-alpha/metabolism
16.
J Immunol ; 187(2): 842-50, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21677141

ABSTRACT

Three surface molecules of mouse CD8(+) dendritic cells (DCs), also found on the equivalent human DC subpopulation, were compared as targets for Ab-mediated delivery of Ags, a developing strategy for vaccination. For the production of cytotoxic T cells, DEC-205 and Clec9A, but not Clec12A, were effective targets, although only in the presence of adjuvants. For Ab production, however, Clec9A excelled as a target, even in the absence of adjuvant. Potent humoral immunity was a result of the highly specific expression of Clec9A on DCs, which allowed longer residence of targeting Abs in the bloodstream, prolonged DC Ag presentation, and extended CD4 T cell proliferation, all of which drove highly efficient development of follicular helper T cells. Because Clec9A shows a similar expression pattern on human DCs, it has particular promise as a target for vaccines of human application.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Cytotoxicity Tests, Immunologic , Dendritic Cells/immunology , Immunophenotyping , Lectins, C-Type/metabolism , Receptors, Immunologic/metabolism , Adjuvants, Immunologic/administration & dosage , Animals , Antigen Presentation/genetics , Antigens, CD/genetics , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/metabolism , Cytotoxicity Tests, Immunologic/methods , Dendritic Cells/metabolism , Humans , Immunophenotyping/methods , Lectins, C-Type/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Minor Histocompatibility Antigens , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Immunologic/genetics , Receptors, Mitogen/genetics , Receptors, Mitogen/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Vaccines, DNA/chemical synthesis , Vaccines, DNA/genetics , Vaccines, DNA/immunology
17.
Blood ; 118(9): 2462-72, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21505196

ABSTRACT

To investigate the role of Aire in thymic selection, we examined the cellular requirements for generation of ovalbumin (OVA)-specific CD4 and CD8 T cells in mice expressing OVA under the control of the rat insulin promoter. Aire deficiency reduced the number of mature single-positive OVA-specific CD4(+) or CD8(+) T cells in the thymus, independent of OVA expression. Importantly, it also contributed in 2 ways to OVA-dependent negative selection depending on the T-cell type. Aire-dependent negative selection of OVA-specific CD8 T cells correlated with Aire-regulated expression of OVA. By contrast, for OVA-specific CD4 T cells, Aire affected tolerance induction by a mechanism that operated independent of the level of OVA expression, controlling access of antigen presenting cells to medullary thymic epithelial cell (mTEC)-expressed OVA. This study supports the view that one mechanism by which Aire controls thymic negative selection is by regulating the indirect presentation of mTEC-derived antigens by thymic dendritic cells. It also indicates that mTECs can mediate tolerance by direct presentation of Aire-regulated antigens to both CD4 and CD8 T cells.


Subject(s)
Antigen Presentation , Antigens/metabolism , Clonal Deletion/immunology , Dendritic Cells/immunology , Epithelial Cells/immunology , Immune Tolerance/immunology , Thymus Gland/immunology , Transcription Factors/immunology , Animals , Antigens/immunology , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Crosses, Genetic , Cytokines/metabolism , Gene Expression Regulation/immunology , Insulin/genetics , Mice , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology , Ovalbumin/metabolism , Promoter Regions, Genetic , Radiation Chimera , Recombinant Fusion Proteins/physiology , Thymus Gland/cytology , Transcription Factors/deficiency , Transcription Factors/genetics , AIRE Protein
18.
J Exp Med ; 207(12): 2703-17, 2010 Nov 22.
Article in English | MEDLINE | ID: mdl-20975040

ABSTRACT

Polyinosinic:polycytidylic acid (poly IC), a double-stranded RNA, is an effective adjuvant in vivo. IFN-λs (also termed IL-28/29) are potent immunomodulatory and antiviral cytokines. We demonstrate that poly IC injection in vivo induces large amounts of IFN-λ, which depended on hematopoietic cells and the presence of TLR3 (Toll-like receptor 3), IRF3 (IFN regulatory factor 3), IRF7, IFN-I receptor, Fms-related tyrosine kinase 3 ligand (FL), and IRF8 but not on MyD88 (myeloid differentiation factor 88), Rig-like helicases, or lymphocytes. Upon poly IC injection in vivo, the IFN-λ production by splenocytes segregated with cells phenotypically resembling CD8α(+) conventional dendritic cells (DCs [cDCs]). In vitro experiments revealed that CD8α(+) cDCs were the major producers of IFN-λ in response to poly IC, whereas both CD8α(+) cDCs and plasmacytoid DCs produced large amounts of IFN-λ in response to HSV-1 or parapoxvirus. The nature of the stimulus and the cytokine milieu determined whether CD8α(+) cDCs produced IFN-λ or IL-12p70. Human DCs expressing BDCA3 (CD141), which is considered to be the human counterpart of murine CD8α(+) DCs, also produced large amounts of IFN-λ upon poly IC stimulation. Thus, IFN-λ production in response to poly IC is a novel function of mouse CD8α(+) cDCs and their human equivalents.


Subject(s)
Antigens, Surface/analysis , CD8 Antigens/analysis , Cytokines/biosynthesis , Dendritic Cells/immunology , Interferon Inducers/pharmacology , Interleukins/biosynthesis , Poly I-C/pharmacology , Animals , Herpesvirus 2, Human , Humans , Interferon Regulatory Factor-3/physiology , Interferon Regulatory Factor-7/physiology , Interferon Regulatory Factors/physiology , Interferons , Interleukin-12/biosynthesis , Mice , Parapoxvirus/immunology , Thrombomodulin , Toll-Like Receptor 3/physiology
19.
Eur J Immunol ; 40(6): 1674-81, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20391433

ABSTRACT

Despite extensive evidence that Plasmodium species are capable of stimulating the immune system, the association of malaria with a higher incidence of other infectious diseases and reduced responses to vaccination against unrelated pathogens suggests the existence of immune suppression. Recently, we provided evidence that blood-stage Plasmodium berghei infection leads to suppression of MHC class I-restricted immunity to third party (non-malarial) antigens as a consequence of systemic DC activation. This earlier study did not, however, determine whether reactivity was also impaired to MHC class II-restricted third party antigens or to Plasmodium antigens themselves. Here, we show that while P. berghei-expressed antigens were presented early in infection, there was a rapid decline in presentation within 4 days, paralleling impairment in MHC class I- and II-restricted presentation of third party antigens. This provides important evidence that P. berghei not only causes immunosuppression to subsequently encountered third party antigens, but also rapidly limits the capacity to generate effective parasite-specific immunity.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Immune Tolerance/immunology , Malaria/immunology , Animals , Antigens, Protozoan/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Parasites/immunology , Plasmodium berghei/immunology
20.
J Immunol ; 184(5): 2243-6, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20124105

ABSTRACT

Despite its potential for involvement in viral immunity, little evidence links TLR3 to adaptive antiviral responses. Here we show that TLR3 is required for the generation of CD8 T cell immunity to HSV-1. The magnitude of the gB-specific CD8 T cell response after flank infection by HSV-1 was significantly reduced in mice lacking TIR domain-containing adaptor-inducing IFN-beta or TLR3, but not MyD88. Impaired CTL induction was evident in chimeric mice lacking TLR3 in bone marrow (BM)-derived cells. Among the dendritic cell subsets, TLR3 was expressed by CD8alpha(+) dendritic cells, known to be involved in priming HSV-1-specific CD8 T cells. Use of mixed BM chimeras revealed that TLR3 and the MHC class I-restriction element must be expressed by the same BM-derived cell for effective priming. These data imply that a cognate linkage between TLR3 and MHC class I is required for efficient CTL priming to HSV-1.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Toll-Like Receptor 3/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Flow Cytometry , H-2 Antigens/genetics , H-2 Antigens/immunology , H-2 Antigens/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/growth & development , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL