Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Immunity ; 54(11): 2481-2496.e6, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34534438

ABSTRACT

How T cell receptor (TCR) signal strength modulates T cell function and to what extent this is modified by immune checkpoint blockade (ICB) are key questions in immunology. Using Nr4a3-Tocky mice, we characterized early quantitative and qualitative changes that occur in CD4+ T cells in relation to TCR signaling strength. We captured how dose- and time-dependent programming of distinct co-inhibitory receptors rapidly recalibrates T cell activation thresholds and visualized the immediate effects of ICB on T cell re-activation. Our findings reveal that anti-PD1 immunotherapy leads to an increased TCR signal strength. We defined a strong TCR signal metric of five genes upregulated by anti-PD1 in T cells (TCR.strong), which was superior to a canonical T cell activation gene signature in stratifying melanoma patient outcomes to anti-PD1 therapy. Our study therefore reveals how analysis of TCR signal strength-and its manipulation-can provide powerful metrics for monitoring outcomes to immunotherapy.


Subject(s)
Antigens/immunology , Immune Checkpoint Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Gene Expression Regulation , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Proteins/genetics , Lymphocyte Activation , Melanoma/drug therapy , Melanoma/etiology , Melanoma/metabolism , Melanoma/pathology , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , T-Lymphocytes/drug effects
2.
Nat Immunol ; 16(8): 819-828, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26147686

ABSTRACT

Fat-associated lymphoid clusters (FALCs) are a type of lymphoid tissue associated with visceral fat. Here we found that the distribution of FALCs was heterogeneous, with the pericardium containing large numbers of these clusters. FALCs contributed to the retention of B-1 cells in the peritoneal cavity through high expression of the chemokine CXCL13, and they supported B cell proliferation and germinal center differentiation during peritoneal immunological challenges. FALC formation was induced by inflammation, which triggered the recruitment of myeloid cells that expressed tumor-necrosis factor (TNF) necessary for signaling via the TNF receptors in stromal cells. Natural killer T cells (NKT cells) restricted by the antigen-presenting molecule CD1d were likewise required for the inducible formation of FALCs. Thus, FALCs supported and coordinated the activation of innate B cells and T cells during serosal immune responses.


Subject(s)
Inflammation/immunology , Intra-Abdominal Fat/immunology , Lymphocytes/immunology , Lymphoid Tissue/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/immunology , Chemokine CXCL13/metabolism , Flow Cytometry , Gene Expression/immunology , Inflammation/genetics , Inflammation/metabolism , Intra-Abdominal Fat/metabolism , Lymphocytes/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Myeloid Cells/immunology , Myeloid Cells/metabolism , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/immunology , Stromal Cells/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
3.
Immunity ; 42(4): 627-39, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25862089

ABSTRACT

Migratory non-lymphoid tissue dendritic cells (NLT-DCs) transport antigens to lymph nodes (LNs) and are required for protective immune responses in the context of inflammation and to promote tolerance to self-antigens in steady-state. However, the molecular mechanisms that elicit steady-state NLT-DC maturation and migration are unknown. By comparing the transcriptome of NLT-DCs in the skin with their migratory counterparts in draining LNs, we have identified a novel NF-κB-regulated gene network specific to migratory DCs. We show that targeted deletion of IKKß in DCs, a major activator of NF-κB, prevents NLT-DC accumulation in LNs and compromises regulatory T cell conversion in vivo. This was associated with impaired tolerance and autoimmunity. NF-κB is generally considered the prototypical pro-inflammatory transcription factor, but this study describes a role for NF-κB signaling in DCs for immune homeostasis and tolerance that could have implications in autoimmune diseases and immunity.


Subject(s)
Dendritic Cells/immunology , Gene Regulatory Networks/immunology , Homeostasis/immunology , Immune Tolerance , NF-kappa B/immunology , Signal Transduction/immunology , Animals , Autoantigens/genetics , Autoantigens/immunology , Autoimmunity , Cell Movement , Dendritic Cells/cytology , Gene Expression Profiling , Gene Expression Regulation , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Knockout , Microarray Analysis , NF-kappa B/genetics , Skin/cytology , Skin/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
4.
J Immunol ; 205(3): 708-719, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32591391

ABSTRACT

Clearance of intracellular infections caused by Salmonella Typhimurium (STm) requires IFN-γ and the Th1-associated transcription factor T-bet. Nevertheless, whereas IFN-γ-/- mice succumb rapidly to STm infections, T-bet-/- mice do not. In this study, we assess the anatomy of immune responses and the relationship with bacterial localization in the spleens and livers of STm-infected IFN-γ-/- and T-bet-/- mice. In IFN-γ-/- mice, there is deficient granuloma formation and inducible NO synthase (iNOS) induction, increased dissemination of bacteria throughout the organs, and rapid death. The provision of a source of IFN-γ reverses this, coincident with subsequent granuloma formation and substantially extends survival when compared with mice deficient in all sources of IFN-γ. T-bet-/- mice induce significant levels of IFN-γ- after challenge. Moreover, T-bet-/- mice have augmented IL-17 and neutrophil numbers, and neutralizing IL-17 reduces the neutrophilia but does not affect numbers of bacteria detected. Surprisingly, T-bet-/- mice exhibit surprisingly wild-type-like immune cell organization postinfection, including extensive iNOS+ granuloma formation. In wild-type mice, most bacteria are within iNOS+ granulomas, but in T-bet-/- mice, most bacteria are outside these sites. Therefore, Th1 cells act to restrict bacteria within IFN-γ-dependent iNOS+ granulomas and prevent dissemination.


Subject(s)
Granuloma/immunology , Nitric Oxide Synthase Type II/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , T-Box Domain Proteins/deficiency , Th1 Cells/immunology , Animals , Granuloma/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Salmonella Infections/genetics , Salmonella typhimurium/genetics , T-Box Domain Proteins/immunology
5.
Blood ; 133(6): 600-604, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30401709

ABSTRACT

Thrombosis is a frequent, life-threatening complication of systemic infection associated with multiple organ damage. We have previously described a novel mechanism of inflammation-driven thrombosis induced by Salmonella Typhimurium infection of mice. Thrombosis in the liver develops 7 days after infection, persisting after the infection resolves, and is monocytic cell dependent. Unexpectedly, thrombosis was not prominent in the spleen at this time, despite carrying a similar bacterial burden as the liver. In this study, we show that thrombosis does occur in the spleen but with strikingly accelerated kinetics compared with the liver, being evident by 24 hours and resolving rapidly thereafter. The distinct kinetics of thrombosis and bacterial burden provides a test of the hypothesis that thrombi form in healthy vessels to trap or remove bacteria from the circulation, often termed immunothrombosis. Remarkably, despite bacteria being detected throughout infected spleens and livers in the early days of infection, immunohistological analysis of tissue sections show that thrombi contain very low numbers of bacteria. In contrast, bacteria are present throughout platelet aggregates induced by Salmonella in vitro. Therefore, we show that thrombosis develops with organ-specific kinetics and challenge the universality of immunothrombosis as a mechanism to capture bacteria in vivo.


Subject(s)
Liver/microbiology , Salmonella Infections/complications , Salmonella typhimurium/pathogenicity , Spleen/microbiology , Thrombosis/microbiology , Animals , Liver/immunology , Liver/pathology , Mice , Mice, Inbred C57BL , Salmonella Infections/microbiology , Spleen/immunology , Spleen/pathology , Thrombosis/immunology , Thrombosis/pathology
6.
Eur J Immunol ; 45(8): 2299-311, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26036767

ABSTRACT

Soluble flagellin (sFliC) from Salmonella Typhimurium (STm) can induce a Th2 response to itself and coadministered antigens through ligation of TLR5. These properties suggest that sFliC could potentially modulate responses to Th1 antigens like live STm if both antigens are given concurrently. After coimmunization of mice with sFliC and STm there was a reduction in Th1 T cells (T-bet(+) IFN-γ(+) CD4 T cells) compared to STm alone and there was impaired clearance of STm. In contrast, there was no significant defect in the early extrafollicular B-cell response to STm. These effects are dependent upon TLR5 and flagellin expression by STm. The mechanism for these effects is not related to IL-4 induced to sFliC but rather to the effects of sFliC coimmunization on DCs. After coimmunization with STm and sFliC, splenic DCs had a lower expression of costimulatory molecules and profoundly altered kinetics of IL-12 and TNFα expression. Ex vivo experiments using in vivo conditioned DCs confirmed the effects of sFliC were due to altered DC function during a critical window in the coordinated interplay between DCs and naïve T cells. This has marked implications for understanding how limits in Th1 priming can be achieved during infection-induced, Th1-mediated inflammation.


Subject(s)
Cytokines/immunology , Dendritic Cells/immunology , Flagellin/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Th1 Cells/immunology , Animals , Cytokines/genetics , Dendritic Cells/pathology , Flagellin/genetics , Immunization , Mice , Mice, Knockout , Salmonella Infections/genetics , Salmonella Infections/prevention & control , Salmonella typhimurium/genetics , Th1 Cells/pathology , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/immunology
7.
Eur J Immunol ; 44(8): 2318-30, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24825601

ABSTRACT

The generation of immune cells from BM precursors is a carefully regulated process. This is essential to limit the potential for oncogenesis and autoimmunity yet protect against infection. How infection modulates this is unclear. Salmonella can colonize systemic sites including the BM and spleen. This resolving infection has multiple IFN-γ-mediated acute and chronic effects on BM progenitors, and during the first week of infection IFN-γ is produced by myeloid, NK, NKT, CD4(+) T cells, and some lineage-negative cells. After infection, the phenotype of BM progenitors rapidly but reversibly alters, with a peak ∼ 30-fold increase in Sca-1(hi) progenitors and a corresponding loss of Sca-1(lo/int) subsets. Most strikingly, the capacity of donor Sca-1(hi) cells to reconstitute an irradiated host is reduced; the longer donor mice are exposed to infection, and Sca-1(hi) c-kit(int) cells have an increased potential to generate B1a-like cells. Thus, Salmonella can have a prolonged influence on BM progenitor functionality not directly related to bacterial persistence. These results reflect changes observed in leucopoiesis during aging and suggest that BM functionality can be modulated by life-long, periodic exposure to infection. Better understanding of this process could offer novel therapeutic opportunities to modulate BM functionality and promote healthy aging.


Subject(s)
Bone Marrow Cells/immunology , Salmonella Infections, Animal/immunology , Stem Cells/immunology , Animals , Antigens, Ly/immunology , Bone Marrow Cells/microbiology , Bone Marrow Cells/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Homeostasis/immunology , Interferon-gamma/immunology , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Salmonella/immunology , Salmonella Infections, Animal/pathology , Stem Cells/microbiology , Stem Cells/pathology
8.
J Immunol ; 189(12): 5745-54, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23152564

ABSTRACT

Mucosal immunity is poorly activated after systemic immunization with protein Ags. Nevertheless, induction of mucosal immunity in such a manner would be an attractive and simple way to overcome the intrinsic difficulties in delivering Ag to such sites. Flagellin from Salmonella enterica serovar Typhimurium (FliC) can impact markedly on host immunity, in part via its recognition by TLR5. In this study, we show that systemic immunization with soluble FliC (sFliC) drives distinct immune responses concurrently in the mesenteric lymph nodes (MLN) and the spleen after i.p. and s.c. immunization. In the MLN, but not the spleen, sFliC drives a TLR5-dependent recruitment of CD103(+) dendritic cells (DCs), which correlates with a diminution in CD103(+) DC numbers in the lamina propria. In the MLN, CD103(+) DCs carry Ag and are the major primers of endogenous and transgenic T cell priming. A key consequence of these interactions with CD103(+) DCs in the MLN is an increase in local regulatory T cell differentiation. In parallel, systemic sFliC immunization results in a pronounced switching of FliC-specific B cells to IgA in the MLN but not elsewhere. Loss of TLR5 has more impact on MLN than splenic Ab responses, reflected in an ablation of IgA, but not IgG, serum Ab titers. Therefore, systemic sFliC immunization targets CD103(+) DCs and drives distinct mucosal T and B cell responses. This offers a potential "Trojan horse" approach to modulate mucosal immunity by systemically immunizing with sFliC.


Subject(s)
Antigens, CD/biosynthesis , Dendritic Cells/immunology , Flagellin/administration & dosage , Flagellin/immunology , Forkhead Transcription Factors/biosynthesis , Immunoglobulin A/biosynthesis , Integrin alpha Chains/biosynthesis , Lymph Nodes/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Coculture Techniques , Dendritic Cells/metabolism , Forkhead Transcription Factors/physiology , Immunization , Lymph Nodes/cytology , Lymph Nodes/metabolism , Mesentery , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mucous Membrane/cytology , Mucous Membrane/immunology , Mucous Membrane/metabolism , Salmonella Infections/metabolism , Salmonella Infections/pathology , Salmonella Infections/prevention & control , Salmonella typhimurium , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Regulatory/metabolism
9.
J Immunol ; 189(5): 2094-8, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22855716

ABSTRACT

Phylogeny shows that CD4 T cell memory and lymph nodes coevolved in placental mammals. In ontogeny, retinoic acid orphan receptor (ROR)γ-dependent lymphoid tissue inducer (LTi) cells program the development of mammalian lymph nodes. In this study, we show that although primary CD4 T cell expansion is normal in RORγ-deficient mice, the persistence of memory CD4 T cells is RORγ-dependent. Furthermore, using bone marrow chimeric mice we demonstrate that LTi cells are the key RORγ-expressing cell type sufficient for memory CD4 T cell survival in the absence of persistent Ag. This effect was specific for CD4 T cells, as memory CD8 T cells survived equally well in the presence or absence of LTi cells. These data demonstrate a novel role for LTi cells, archetypal members of the innate lymphoid cell family, in supporting memory CD4 T cell survival in vivo.


Subject(s)
Immunologic Memory , Lymphoid Tissue/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adaptive Immunity/genetics , Animals , Cell Death/genetics , Cell Death/immunology , Cell Survival/genetics , Cell Survival/immunology , Immunity, Innate/genetics , Immunologic Memory/genetics , Lymphoid Tissue/cytology , Lymphoid Tissue/transplantation , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/deficiency , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Radiation Chimera/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/pathology
10.
J Immunol ; 189(12): 5527-32, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23162127

ABSTRACT

Vaccination with purified capsular polysaccharide Vi Ag from Salmonella typhi can protect against typhoid fever, although the mechanism for its efficacy is not clearly established. In this study, we have characterized the B cell response to this vaccine in wild-type and T cell-deficient mice. We show that immunization with typhoid Vi polysaccharide vaccine rapidly induces proliferation in B1b peritoneal cells, but not in B1a cells or marginal zone B cells. This induction of B1b proliferation is concomitant with the detection of splenic Vi-specific Ab-secreting cells and protective Ab in Rag1-deficient B1b cell chimeras generated by adoptive transfer-induced specific Ab after Vi immunization. Furthermore, Ab derived from peritoneal B cells is sufficient to confer protection against Salmonella that express Vi Ag. Expression of Vi by Salmonella during infection did not inhibit the development of early Ab responses to non-Vi Ags. Despite this, the protection conferred by immunization of mice with porin proteins from Salmonella, which induce Ab-mediated protection, was reduced postinfection with Vi-expressing Salmonella, although protection was not totally abrogated. This work therefore suggests that, in mice, B1b cells contribute to the protection induced by Vi Ag, and targeting non-Vi Ags as subunit vaccines may offer an attractive strategy to augment current Vi-based vaccine strategies.


Subject(s)
Antigens, Bacterial/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Polysaccharides, Bacterial/biosynthesis , Salmonella typhi/immunology , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/physiology , Antigens, Bacterial/biosynthesis , B-Lymphocyte Subsets/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneal Cavity/cytology , Peritoneal Cavity/microbiology , Peritoneum/cytology , Peritoneum/immunology , Peritoneum/metabolism , Polysaccharides, Bacterial/immunology , Porins , Salmonella Vaccines/administration & dosage , Salmonella Vaccines/biosynthesis , Salmonella Vaccines/immunology , Typhoid Fever/immunology , Typhoid Fever/metabolism , Typhoid Fever/prevention & control
11.
J Immunol ; 189(9): 4266-74, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22993205

ABSTRACT

Thymic atrophy is a frequent consequence of infection with bacteria, viruses, and parasites and is considered a common virulence trait between pathogens. Multiple reasons have been proposed to explain this atrophy, including premature egress of immature thymocytes, increased apoptosis, or thymic shutdown to prevent tolerance to the pathogen from developing. The severe loss in thymic cell number can reflect an equally dramatic reduction in thymic output, potentially reducing peripheral T cell numbers. In this study, we examine the relationship between systemic Salmonella infection and thymic function. During infection, naive T cell numbers in peripheral lymphoid organs increase. Nevertheless, this occurs despite a pronounced thymic atrophy caused by viable bacteria, with a peak 50-fold reduction in thymocyte numbers. Thymic atrophy is not dependent upon homeostatic feedback from peripheral T cells or on regulation of endogenous glucocorticoids, as demonstrated by infection of genetically altered mice. Once bacterial numbers fall, thymocyte numbers recover, and this is associated with increases in the proportion and proliferation of early thymic progenitors. During atrophy, thymic T cell maturation is maintained, and single-joint TCR rearrangement excision circle analysis reveals there is only a modest fall in recent CD4(+) thymic emigrants in secondary lymphoid tissues. Thus, thymic atrophy does not necessarily result in a matching dysfunctional T cell output, and thymic homeostasis can constantly adjust to systemic infection to ensure that naive T cell output is maintained.


Subject(s)
Recovery of Function/immunology , Salmonella Infections/immunology , Thymus Gland/immunology , Thymus Gland/pathology , Animals , Atrophy , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Cell Differentiation/immunology , Cell Movement/immunology , Mice , Salmonella Infections/pathology , Salmonella Infections/physiopathology , Salmonella typhimurium/immunology , Thymus Gland/microbiology
12.
J Immunol ; 186(9): 5406-12, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21451112

ABSTRACT

TLR5-deficient mice have been reported to develop spontaneous intestinal inflammation and metabolic abnormalities. However, we report that TLR5-deficient mice from two different animal colonies display no evidence of basal inflammatory disease, metabolic abnormalities, or enhanced resistance to Salmonella infection. In contrast, the absence of TLR5 hindered the initial activation and clonal expansion of intestinal flagellin-specific CD4 T cells following oral Salmonella infection. Together, these data demonstrate that a basal inflammatory phenotype is not a consistent feature of TLR5-deficient mice and document a novel role for TLR5 in the rapid targeting of flagellin by intestinal pathogen-specific CD4 T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Flagellin/immunology , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/metabolism , Toll-Like Receptor 5/deficiency , Animals , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Salmonella Infections, Animal/pathology , Toll-Like Receptor 5/immunology
13.
J Immunol ; 187(4): 1553-65, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21734076

ABSTRACT

Hematopoietic cells constitutively express CD31/PECAM1, a signaling adhesion receptor associated with controlling responses to inflammatory stimuli. Although expressed on CD4(+) T cells, its function on these cells is unclear. To address this, we have used a model of systemic Salmonella infection that induces high levels of T cell activation and depends on CD4(+) T cells for resolution. Infection of CD31-deficient (CD31KO) mice demonstrates that these mice fail to control infection effectively. During infection, CD31KO mice have diminished numbers of total CD4(+) T cells and IFN-γ-secreting Th1 cells. This is despite a higher proportion of CD31KO CD4(+) T cells exhibiting an activated phenotype and an undiminished capacity to prime normally and polarize to Th1. Reduced numbers of T cells reflected the increased propensity of naive and activated CD31KO T cells to undergo apoptosis postinfection compared with wild-type T cells. Using adoptive transfer experiments, we show that loss of CD31 on CD4(+) T cells alone is sufficient to account for the defective CD31KO T cell accumulation. These data are consistent with CD31 helping to control T cell activation, because in its absence, T cells have a greater propensity to become activated, resulting in increased susceptibility to become apoptotic. The impact of CD31 loss on T cell homeostasis becomes most pronounced during severe, inflammatory, and immunological stresses such as those caused by systemic Salmonella infection. This identifies a novel role for CD31 in regulating CD4 T cell homeostasis.


Subject(s)
Apoptosis/immunology , Lymphocyte Activation/immunology , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Salmonella Infections/immunology , Salmonella/immunology , Th1 Cells/immunology , Adoptive Transfer , Animals , Apoptosis/genetics , Cell Survival/genetics , Cell Survival/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Lymphocyte Activation/genetics , Mice , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Salmonella/genetics , Salmonella Infections/genetics
14.
Eur J Immunol ; 41(9): 2654-65, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21630252

ABSTRACT

Control of intracellular Salmonella infection requires Th1 priming and IFN-γ production. Here, we show that efficient Th1 priming after Salmonella infection requires CD11c(+) CD11b(hi) F4/80(+) monocyte-derived dendritic cells (moDCs). In non-infected spleens, moDCs are absent from T-cell zones (T zones) of secondary lymphoid tissues, but by 24 h post-infection moDCs are readily discernible in these sites. The accumulation of moDCs is more dependent upon bacterial viability than bacterial virulence. Kinetic studies showed that moDCs were necessary to prime but not sustain Th1 responses, while ex vivo studies showed that antigen-experienced moDCs were sufficient to induce T-cell proliferation and IFN-γ production via a TNF-α-dependent mechanism. Importantly, moDCs and cDCs when co-cultured induced superior Th1 differentiation than either subset alone, and this activity was independent of TNF-α. Thus, optimal Th1 development to Salmonella requires the rapid accumulation of moDCs within T zones and their collaboration with cDCs.


Subject(s)
Dendritic Cells/metabolism , Salmonella Infections/immunology , Salmonella/immunology , Spleen/pathology , Th1 Cells/metabolism , Animals , Antigen Presentation , Antigens, Differentiation/biosynthesis , CD11b Antigen/biosynthesis , CD11c Antigen/biosynthesis , Cell Communication , Cell Differentiation , Cell Proliferation , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/microbiology , Dendritic Cells/pathology , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Monocytes/pathology , Salmonella/pathogenicity , Th1 Cells/immunology , Th1 Cells/microbiology , Th1 Cells/pathology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
15.
Eur J Immunol ; 41(6): 1606-18, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21469112

ABSTRACT

Clearance of disseminated Salmonella infection requires bacterial-specific Th1 cells and IFN-γ production, and Th1-promoting vaccines are likely to help control these infections. Consequently, vaccine design has focused on developing Th1-polarizing adjuvants or Ag that naturally induce Th1 responses. In this study, we show that, in mice, immunization with soluble, recombinant FliC protein flagellin (sFliC) induces Th2 responses as evidenced by Ag-specific GATA-3, IL-4 mRNA, and protein induction in CD62L(lo) CD4(+) T cells without associated IFN-γ production. Despite these Th2 features, sFliC immunization can enhance the development of protective Th1 immunity during subsequent Salmonella infection in an Ab-independent, T-cell-dependent manner. Salmonella infection in sFliC-immunized mice resulted in augmented Th1 responses, with greater bacterial clearance and increased numbers of IFN-γ-producing CD4(+) T cells, despite the early induction of Th2 features to sFliC. The augmented Th1 immunity after sFliC immunization was regulated by T-bet although T-bet is dispensable for primary responses to sFliC. These findings show that there can be flexibility in T-cell responses to some subunit vaccines. These vaccines may induce Th2-type immunity during primary immunization yet promote Th1-dependent responses during later infection. This suggests that designing Th1-inducing subunit vaccines may not always be necessary since this can occur naturally during subsequent infection.


Subject(s)
Bacterial Vaccines , Flagellin/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism , Animals , Bacterial Load , Cells, Cultured , Gene Expression Regulation , Immunization , Interferon-gamma/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Salmonella Infections/microbiology , Salmonella typhimurium/growth & development , Salmonella typhimurium/pathogenicity , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Cell Antigen Receptor Specificity , Th1 Cells/immunology , Th1 Cells/microbiology , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/microbiology , Th2 Cells/pathology
16.
Immunol Invest ; 41(1): 104-16, 2012.
Article in English | MEDLINE | ID: mdl-21696341

ABSTRACT

The intestinal mucosa is exposed to a vast antigenic contact. Several antigen presenting cell (APCs) have been described within the gut associated lymphoid tissue (GALT) (Peyer's patches, lamina propria, mesenteric lymph nodes, muscular layer); however, this has been done almost exclusively in adult organisms. As there is no characterization of intestinal muscular layer's APCs during early neonate development we adapted the conventional technique used in adults, to the neonate intestine. We obtained the intestinal muscular layer from early neonates (days 0-3 upon birth) and from young mice (2 and 3 weeks after birth). A planar network of CD45(+), MHC-II(+), DEC-205(+) cells with irregular, some with prominent dendritic morphology was found at birth under basal physiological conditions, whereas Langerin(+) DCs appeared after two weeks. The variations seen in CD45(+), MHC-II(+) and DEC-205(+) cells along the early neonatal development, could be related to the new challenges by intestinal antigen exposure from the newborn diet (breast milk, solid food), and to important environmental changes (start walking, exploring the surroundings, etc). Our study reveals the presence of APCs in intestinal muscular layer at birth, and their subsequent changes in physiological, non-induced conditions, contributing basic information about these cells in the neonate intestinal immune system.


Subject(s)
Antigen-Presenting Cells/metabolism , Biomarkers/metabolism , Intestinal Mucosa/immunology , Intestine, Small/pathology , Adult , Animals , Animals, Newborn , Antigen-Presenting Cells/pathology , Antigens, CD/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Immunohistochemistry , Intestinal Mucosa/pathology , Lectins, C-Type/metabolism , Leukocyte Common Antigens/metabolism , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred BALB C , Minor Histocompatibility Antigens , Muscles/pathology , Receptors, Cell Surface/metabolism
17.
Infect Immun ; 79(11): 4342-52, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21859856

ABSTRACT

Salmonella enterica is a major cause of morbidity worldwide and mortality in children and immunocompromised individuals in sub-Saharan Africa. Outer membrane proteins of Salmonella are of significance because they are at the interface between the pathogen and the host, they can contribute to adherence, colonization, and virulence, and they are frequently targets of antibody-mediated immunity. In this study, the properties of SadA, a purported trimeric autotransporter adhesin of Salmonella enterica serovar Typhimurium, were examined. We demonstrated that SadA is exposed on the Salmonella cell surface in vitro and in vivo during infection of mice. Expression of SadA resulted in cell aggregation, biofilm formation, and increased adhesion to human intestinal Caco-2 epithelial cells. Immunization of mice with folded, full-length, purified SadA elicited an IgG response which provided limited protection against bacterial challenge. When anti-SadA IgG titers were enhanced by administering alum-precipitated protein, a modest additional protection was afforded. Therefore, despite SadA having pleiotropic functions, it is not a dominant, protective antigen for antibody-mediated protection against Salmonella.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion/physiology , Biofilms , Gene Expression Regulation, Bacterial/physiology , Membrane Proteins/metabolism , Salmonella typhimurium/metabolism , Adhesins, Bacterial/genetics , Alum Compounds , Animals , Bacterial Adhesion/genetics , Caco-2 Cells , Escherichia coli K12/metabolism , Humans , Immunoglobulin G , Membrane Proteins/genetics , Mice , Phylogeny , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity , Virulence
18.
J Immunol ; 182(3): 1305-13, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155476

ABSTRACT

Mice lacking complement components show delayed development of prion disease following peripheral inoculation. The delay could relate to reduced scrapie prion protein (PrP(Sc)) accumulation on follicular dendritic cells (DCs). However conventional DCs (cDCs) play a crucial role in the early pathogenesis of prion diseases and complement deficiency could result in decreased PrP(Sc) uptake by cDCs in the periphery. To explore this possibility, we cultured murine splenic or gut-associated lymph node cDCs with scrapie-infected whole brain homogenate in the presence or absence of complement. Uptake decreased significantly if the serum in the cultures was heat-inactivated. Because heat inactivation primarily denatures C1q, we used serum from C1q(-/-) mice and showed that PrP(Sc) uptake was markedly decreased. PrP(Sc) internalization was saturable and temperature-dependent, suggesting receptor-mediated uptake. Furthermore, uptake characteristics differed from fluid-phase endocytosis. Immunofluorescence showed colocalization of C1q and PrP(Sc), suggesting interaction between these molecules. We evaluated the expression of several complement receptors on cDCs and confirmed that cDCs that take up PrP(Sc) express one of the C1q receptors, calreticulin. Our results show that C1q participates in PrP(Sc) uptake by cDCs, revealing a critical role for cDCs in initial prion capture, an event that takes place before the PrP(Sc) accumulation within the follicular DC network.


Subject(s)
Complement C1q/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , PrPSc Proteins/metabolism , Scrapie/immunology , Scrapie/metabolism , Animals , Brain/cytology , Brain/immunology , Brain/metabolism , Cells, Cultured , Coculture Techniques , Complement C1q/deficiency , Complement C1q/genetics , Dendritic Cells/pathology , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Endocytosis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Transport/immunology , Receptors, Complement/biosynthesis , Scrapie/pathology
19.
J Immunol Res ; 2021: 5511841, 2021.
Article in English | MEDLINE | ID: mdl-33997054

ABSTRACT

Dengue is a worldwide expanding threat caused by dengue virus (DENV) infection. To date, no specific treatment or effective vaccine is available. Antibodies produced by plasma cells (PCs) might be involved concomitantly in protection and severe dengue immunopathology. Although a massive appearance of PCs has been reported during acute DENV infection in humans, this response has been poorly characterized. Here, we show the dynamic of PC generation in immune-competent mice cutaneously inoculated with DENV compared with two control experimental groups: mice inoculated with inactivated DENV or with PBS. We found that PC numbers increased significantly in the skin-draining lymph node (DLN), peaking at day 10 and abruptly decreasing by day 14 after DENV inoculation. Class-switched IgG+ PCs appeared from day 7 and dominated the response, while in contrast, the frequency of IgM+ PCs decreased from day 7 onwards. Even though the kinetic of the response was similar between DENV- and iDENV-inoculated mice, the intensity of the response was significantly different. Interestingly, we demonstrated a similar PC response to virus antigens (E and prM) by ELISPOT. In situ characterization showed that PCs were distributed in the medullary cords and in close proximity to germinal centers (GCs), suggesting both an extrafollicular and a GC origin. Proliferating PCs (Ki-67+) were found as early as 3-day postinoculation, and in-depth analysis showed that these PCs were in active phases of cell cycle during the kinetic. Finally, we found a progressive appearance of high-affinity neutralizing DENV-specific IgG further supporting GC involvement. Of note, these antibodies seem to be highly cross-reactive, as a large proportion recognizes Zika virus (ZIKV). The strong PC response to skin-inoculated DENV in this work resembles the findings already described in humans. We consider that this study contributes to the understanding of the in vivo biology of the humoral immune response to DENV in an immunocompetent murine model.


Subject(s)
Dengue Virus/immunology , Dengue/immunology , Plasma Cells/immunology , Animals , Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/metabolism , Antibodies, Viral/analysis , Antibodies, Viral/metabolism , Cross Reactions , Dengue/pathology , Dengue/virology , Disease Models, Animal , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Humans , Male , Mice , Plasma Cells/metabolism , Skin/immunology , Skin/pathology , Skin/virology , Specific Pathogen-Free Organisms , Zika Virus/immunology
20.
Front Immunol ; 11: 744, 2020.
Article in English | MEDLINE | ID: mdl-32395120

ABSTRACT

Antigen capturing at the periphery is one of the earliest, crucial functions of antigen-presenting cells (APCs) to initiate immune responses. Langerhans cells (LCs), the epidermal APCs migrate to draining lymph nodes (DLNs) upon acquiring antigens. An arsenal of endocytic molecules is available to this end, including lectins and pathogen recognition receptors (PRRs). However, cutaneous LCs are poorly defined in the early neonatal period. We assessed endocytic molecules expression in situ: Mannose (CD206)-, Scavenger (SRA/CD204)-, Complement (CD2l, CDllb)-, and Fc-Receptors (CD16/32, CD23) as well as CD1d, CD14, CD205, Langerin (CD207), MHCII, and TLR4 in unperturbed epidermal LCs from both adult and early neonatal mice. As most of these markers were negative at birth (day 0), LC presence was revealed with the conspicuous, epidermal LC-restricted ADPase (and confirmed with CD45) staining detecting that they were as numerous as adult ones. Unexpectedly, most LCs at day 0 expressed CD14 and CD204 while very few were MHCII+ and TLR4+. In contrast, adult LCs lacked all these markers except Langerin, CD205, CD11b, MHCII and TLR4. Intriguingly, the CD204+ and CD14+ LCs predominant at day 0, apparently disappeared by day 4. Upon cutaneous FITC application, LCs were reduced in the skin and a CD204+MHCII+FITC+ population with high levels of CD86 subsequently appeared in DLNs, with a concomitant increased percentage of CD3+CD69+ T cells, strongly suggesting that neonatal LCs were able both to ferry the cutaneous antigen into DLNs and to activate neonatal T cells in vivo. Cell cycle analysis indicated that neonatal T cells in DLNs responded with proliferation. Our study reveals that epidermal LCs are present at birth, but their repertoire of endocytic molecules and PRRs differs to that of adult ones. We believe this to be the first description of CDl4, CD204 and TLR4 in neonatal epidermal LCs in situ. Newborns' LCs express molecules to detect antigens during early postnatal periods, are able to take up local antigens and to ferry them into DLNs conveying the information to responsive neonatal T cells.


Subject(s)
Langerhans Cells/immunology , Langerhans Cells/physiology , Receptors, Cell Surface/metabolism , T-Lymphocytes/metabolism , Animals , Animals, Newborn , Cell Movement , Cell Proliferation , Epidermal Cells/metabolism , Female , Lymph Nodes , Mice , Mice, Inbred BALB C , Pregnancy , Skin/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7
SELECTION OF CITATIONS
SEARCH DETAIL