Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
2.
Blood ; 122(2): 209-18, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23723451

ABSTRACT

B-cell numbers and immunoglobulin (Ig) titers can increase several logs during immune responses. In contrast to this plasticity and despite constant renewal, B-cell numbers are stable in the absence of immunization. We assessed the role of serum Igs in maintaining specific B-cell subset homeostasis at steady state. Using mice genetically deficient in secreted IgM only (secretory µ chain-deficient), in switched Igs and hypermutated IgM (activation-induced cytidine deaminase-deficient), or fully agammaglobulemic (AID(-/-)µS(-/-)), we dissected the contribution of different Ig classes to 4 phenotypes associated with loss of serum Igs: 1) increased splenic B-cell numbers, mostly of the B1 and marginal zone (MZ) B-cell subtypes; 2) enlarged germinal centers (GCs) in spleen and mesenteric lymph nodes; 3) enrichment in IRF4(+)CD138(-) plasmablast-like cells; and 4) overexpression of IgM in several cell subsets. Complementation experiments based on either mixed bone marrow reconstitution of chimeras or Ig infusion, and analysis of mice raised in germ-free conditions reveal a negative feedback mechanism in which MZ and B1 cell numbers are under the control of naturally secreted Igs as the result of an intrinsic property of the immune system, whereas GC development is under indirect control of secreted Igs that limit bacterial species triggering GC reactions.


Subject(s)
Antibody Formation/immunology , B-Lymphocyte Subsets/immunology , Metagenome , Animals , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Female , Germinal Center/immunology , Immunoglobulin M/metabolism , Interferon Regulatory Factors/metabolism , Lymphoid Tissue/immunology , Mice , Mice, Knockout , Plasma Cells/cytology , Plasma Cells/immunology , Plasma Cells/metabolism , Spleen/immunology , Syndecan-1/metabolism
3.
Blood ; 120(23): 4552-9, 2012 Nov 29.
Article in English | MEDLINE | ID: mdl-23065152

ABSTRACT

Antibody-forming cells (AFCs) expressing the chemokine receptor CXCR3 are recruited to sites of inflammation where they help clear pathogens but may participate in autoimmune diseases. Here we identify a mechanism that induces CXCR3 expression by AFC and germinal center (GC) B cells. This happens when CD8 T cells are recruited into CD4 T cell-dependent B-cell responses. Ovalbumin-specific CD4 T cells (OTII) were transferred alone or with ovalbumin-specific CD8 T cells (OTI) and the response to subcutaneous alum-precipitated ovalbumin was followed in the draining lymph nodes. OTII cells alone induce T helper 2-associated class switching to IgG1, but few AFC or GC B cells express CXCR3. By contrast, OTI-derived IFN-γ induces most responding GC B cells and AFCs to express high levels of CXCR3, and diverse switching to IgG2a, IgG2b, with some IgG1. Up-regulation of CXCR3 by GC B cells and AFCs and their migration toward its ligand CXCL10 are shown to depend on B cells' intrinsic T-bet, a transcription factor downstream of the IFN-γR signaling. This model clarifies how precursors of long-lived AFCs and memory B cells acquire CXCR3 that causes their migration to inflammatory foci.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Receptors, CXCR3/immunology , T-Box Domain Proteins/immunology , Vaccines/immunology , Adoptive Transfer , Alum Compounds , Animals , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Flow Cytometry , Germinal Center/immunology , Germinal Center/metabolism , Immunization/methods , Interferon-gamma/immunology , Interferon-gamma/metabolism , Ligands , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Up-Regulation/genetics
4.
Proc Natl Acad Sci U S A ; 107(40): 17292-7, 2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20855629

ABSTRACT

Alum-precipitated protein (alum protein) vaccines elicit long-lasting neutralizing antibody responses that prevent bacterial exotoxins and viruses from entering cells. Typically, these vaccines induce CD4 T cells to become T helper 2 (Th2) cells that induce Ig class switching to IgG1. We now report that CD8 T cells also respond to alum proteins, proliferating extensively and producing IFN-γ, a key Th1 cytokine. These findings led us to question whether adoptive transfer of antigen-specific CD8 T cells alters the characteristic CD4 Th2 response to alum proteins and the switching pattern in responding B cells. To this end, WT mice given transgenic ovalbumin (OVA)-specific CD4 (OTII) or CD8 (OTI) T cells, or both, were immunized with alum-precipitated OVA. Cotransfer of antigen-specific CD8 T cells skewed switching patterns in responding B cells from IgG1 to IgG2a and IgG2b. Blocking with anti-IFN-γ antibody largely inhibited this altered B-cell switching pattern. The transcription factor T-bet is required in B cells for IFN-γ-dependent switching to IgG2a. By contrast, we show that this transcription factor is dispensable in B cells both for IFN-γ-induced switching to IgG2b and for inhibition of switching to IgG1. Thus, T-bet dependence identifies distinct transcriptional pathways in B cells that regulate IFN-γ-induced switching to different IgG isotypes.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunoglobulin Class Switching , Interferon-gamma/immunology , Ovalbumin/immunology , T-Box Domain Proteins/metabolism , Vaccines/immunology , Adoptive Transfer , Alum Compounds , Animals , B-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Mice , Mice, Inbred C57BL
5.
Front Immunol ; 14: 1155200, 2023.
Article in English | MEDLINE | ID: mdl-37063899

ABSTRACT

Introduction: C-type lectin receptor (CLR) agonists emerged as superior inducers of primary B cell responses in early life compared with Toll-like receptor (TLR) agonists, while both types of adjuvants are potent in adults. Methods: Here, we explored the mechanisms accounting for the differences in neonatal adjuvanticity between a CLR-based (CAF®01) and a TLR4-based (GLA-SE) adjuvant administered with influenza hemagglutinin (HA) in neonatal mice, by using transcriptomics and systems biology analyses. Results: On day 7 after immunization, HA/CAF01 increased IL6 and IL21 levels in the draining lymph nodes, while HA/GLA-SE increased IL10. CAF01 induced mixed Th1/Th17 neonatal responses while T cell responses induced by GLA-SE had a more pronounced Th2-profile. Only CAF01 induced T follicular helper (Tfh) cells expressing high levels of IL21 similar to levels induced in adult mice, which is essential for germinal center (GC) formation. Accordingly, only CAF01- induced neonatal Tfh cells activated adoptively transferred hen egg lysozyme (HEL)-specific B cells to form HEL+ GC B cells in neonatal mice upon vaccination with HEL-OVA. Discussion: Collectively, the data show that CLR-based adjuvants are promising neonatal and infant adjuvants due to their ability to harness Tfh responses in early life.


Subject(s)
B-Lymphocytes , Germinal Center , Lectins, C-Type , T Follicular Helper Cells , Animals , Mice , Adjuvants, Immunologic/pharmacology , Lectins, C-Type/agonists , Animals, Newborn
6.
Eur J Immunol ; 41(6): 1573-82, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21469117

ABSTRACT

NF-κB1-dependent signaling directs the development of CD4(+) Th2 cells during allergic airway inflammation and protective responses to helminth infection. Here, we show that IL-4 and IL-13 production is NF-κB1-dependent in mouse OVA-specific CD4(+) (OTII) T cells responding to alum-precipitated OVA (alumOVA) immunization. More surprisingly, we found that NF-κB1 deficiency in OTII cells also selectively impairs their CXCR5 induction by alumOVA without affecting upregulation of BCL6, IL-21, OX40 and CXCR4 mRNA and PD-1 protein. This results in functional impairment of follicular helper T cells. Thus, fewer germinal center B cells develop in LN responses to alumOVA in T-cell-deficient mice reconstituted with NF-κB1(-/-) OTII cells as opposed to NF-κB1(+/+) OTII cells, while plasma cell numbers are comparable. Unlike CXCR5 induction in CD4(+) T cells, NF-κB1-deficient recirculating follicular B cells are shown to express normal levels of CXCR5. The selective effects of NF-κB1-deficiency on Th2 and follicular helper T cell induction do not appear to be due to altered expression of the Th2-associated transcription factors - GATA-3, c-Maf and Ikaros. Altogether, these results suggest that NF-κB1 regulates the expression of CXCR5 on CD4(+) T cells primed in vivo, and thus selectively controls the T-cell-dependent germinal center component of B-cell response to alumOVA.


Subject(s)
B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/metabolism , NF-kappa B/metabolism , Receptors, CXCR5/metabolism , Th2 Cells/metabolism , Adoptive Transfer , Alum Compounds/administration & dosage , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Germinal Center/pathology , Immunization , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/immunology , Ovalbumin/immunology , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , Th2 Cells/immunology , Th2 Cells/pathology , Up-Regulation/genetics
7.
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
8.
Blood ; 116(11): 1867-75, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20538807

ABSTRACT

Long-lived plasma cells in the bone marrow produce memory antibodies that provide immune protection persisting for decades after infection or vaccination but can also contribute to autoimmune and allergic diseases. However, the composition of the microenvironmental niches that are important for the generation and maintenance of these cells is only poorly understood. Here, we demonstrate that, within the bone marrow, plasma cells interact with the platelet precursors (megakaryocytes), which produce the prominent plasma cell survival factors APRIL (a proliferation-inducing ligand) and IL-6 (interleukin-6). Accordingly, reduced numbers of immature and mature plasma cells are found in the bone marrow of mice deficient for the thrombopoietin receptor (c-mpl) that show impaired megakaryopoiesis. After immunization, accumulation of antigen-specific plasma cells in the bone marrow is disturbed in these mice. Vice versa, injection of thrombopoietin allows the accumulation and persistence of a larger number of plasma cells generated in the course of a specific immune response in wild-type mice. These results demonstrate that megakaryocytes constitute an important component of the niche for long-lived plasma cells in the bone marrow.


Subject(s)
Bone Marrow Cells/metabolism , Megakaryocytes/metabolism , Plasma Cells/metabolism , Stem Cell Niche/metabolism , Animals , Bone Marrow Cells/cytology , Cell Communication/drug effects , Cell Count , Cells, Cultured , Female , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Megakaryocytes/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Ovalbumin/immunology , Ovalbumin/pharmacology , Plasma Cells/cytology , Plasma Cells/drug effects , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Niche/cytology , Thrombopoietin/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
9.
Front Immunol ; 13: 873217, 2022.
Article in English | MEDLINE | ID: mdl-35464469

ABSTRACT

Antibody-secreting cells (ASCs) contribute to immunity through production of antibodies and cytokines. Identification of specific markers of ASC would allow selective targeting of these cells in several disease contexts. Here, we performed an unbiased, large-scale protein screening, and identified twelve new molecules that are specifically expressed by murine ASCs. Expression of these markers, particularly CD39, CD81, CD130, and CD326, is stable and offers an improved resolution for ASC identification. We accessed their expression in germ-free conditions and in T cell deficient mice, showing that at least in part their expression is controlled by microbial- and T cell-derived signals. Further analysis of lupus mice revealed the presence of a subpopulation of LAG-3- plasma cells, co-expressing high amounts of CD39 and CD326 in the bone marrow. This population was IgM+ and correlated with IgM anti-dsDNA autoantibodies in sera. Importantly, we found that CD39, CD81, CD130, and CD326 are also expressed by human peripheral blood and bone marrow ASCs. Our data provide innovative insights into ASC biology and function in mice and human, and identify an intriguing BM specific CD39++CD326++ ASC subpopulation in autoimmunity.


Subject(s)
Bone Marrow , Plasma Cells , Animals , Antibodies, Antinuclear , Antibody-Producing Cells , Biomarkers/metabolism , Bone Marrow/metabolism , Humans , Immunoglobulin M , Mice , Plasma Cells/metabolism
10.
Immunology ; 132(1): 123-33, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20875075

ABSTRACT

Acute infection with Trypanosoma cruzi, the aetiological agent of Chagas' disease, results in parasitaemia and polyclonal lymphocyte activation. It has been reported that polyclonal B-cell activation is associated with hypergammaglobulinaemia and delayed parasite-specific antibody response. In the present study we analysed the development of a B-cell response within the different microenvironments of the spleen during acute T. cruzi infection. We observed massive germinal centre (GC) and extrafollicular (EF) responses at the peak of infection. However, the EF foci were evident since day 3 post-infection (p.i.), and, early in the infection, they mainly provided IgM. The EF foci response reached its peak at 11 days p.i. and extended from the red pulp into the periarteriolar lymphatic sheath. The GCs were detected from day 8 p.i. At the peak of parasitaemia, CD138(+) B220(+) plasma cells in EF foci, red pulp and T-cell zone expressed IgM and all the IgG isotypes. Instead of the substantial B-cell response, most of the antibodies produced by splenic cells did not target the parasite, and parasite-specific IgG isotypes could be detected in sera only after 18 days p.i. We also observed that the bone marrow of infected mice presented a strong reduction in CD138(+) B220(+) cells compared with that of normal mice. Hence, in acute infection with T. cruzi, the spleen appears to be the most important lymphoid organ that lodges plasma cells and the main producer of antibodies. The development of a B-cell response during T. cruzi infection shows features that are particular to T. cruzi and other protozoan infection but different to other infections or immunization with model antigens.


Subject(s)
Antibodies/immunology , B-Lymphocytes/immunology , Spleen/cytology , Spleen/immunology , Trypanosoma cruzi/immunology , Animals , Antibodies, Protozoan/immunology , Chagas Disease/immunology , Chagas Disease/parasitology , Lymphocyte Count , Mice , Mice, Inbred BALB C , Trypanosoma cruzi/isolation & purification
11.
J Immunol ; 182(4): 2113-23, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19201864

ABSTRACT

IL-6 and APRIL influence the growth, differentiation, and survival of normal and neoplastic Ab-forming cells (AFC). In this study, we identify two subsets of myeloid cells that associate with the AFC and are the main producers of these factors during a T-dependent Ab response to alum-precipitated protein in mouse lymph nodes. First CD11c(+)CD8alpha(-) dendritic cells located in the perivascular area of the T zone provide about half of the IL-6 mRNA produced in the node together with significant amounts of APRIL mRNA. The number of these cells increases during the response, at least in part due to local proliferation. The second subset comprises Gr1(+)CD11b(+)F4/80(+) monocyte/macrophages. These colonize the medullary cords during the response and are the other main IL-6 mRNA producers and the greatest source of APRIL mRNA. This medullary cord monocyte/macrophage subset results in local increase of APRIL mRNA that mirrors the polarity of CXCL12 expression in the node. The distribution of these myeloid cell subsets correlates with a gradient of AFC maturation assessed by progressive loss of Ki67 as AFC pass from the B cell follicle along the perivascular areas to the medullary cords.


Subject(s)
Dendritic Cells/cytology , Interleukin-6/immunology , Leukocytes, Mononuclear/cytology , Lymph Nodes/cytology , Plasma Cells/cytology , Tumor Necrosis Factor Ligand Superfamily Member 13/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cell Proliferation , Dendritic Cells/immunology , Flow Cytometry , Leukocytes, Mononuclear/immunology , Lymph Nodes/immunology , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microdissection , Microscopy, Confocal , Myeloid Cells/cytology , Myeloid Cells/immunology , Plasma Cells/immunology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
12.
Eur J Immunol ; 39(6): 1573-86, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19462378

ABSTRACT

This study characterizes the diversity of CD4 Th cells produced during a Th2 response in vivo. Kinetics of effector and memory cell differentiation by mouse OVA-specific CD4 T cells was followed during primary responses to alum-precipitated OVA. The complexity of the CD4 T response was assessed in nodes draining and distant from the site of immunization for phenotype, location and function. By 3 days IL-4-producing effector CD4 T cells developed in the draining node and a proportion of the responding cells had migrated to B-cell follicles, while yet others had left the draining node. Some of these early migrant cells were recirculating cells with a central memory phenotype. These had divided four or more times in the draining node before migrating to distant nodes not exposed to antigen. We questioned the responsiveness of these early central-memory-like cells on secondary antigen challenge at sites distant to the primary immunization. They re-entered cell cycle and migrated to B-cell follicles, much more rapidly than naive CD4 T cells and could still be induced to produce IL-4. Their production and survival were independent of the starting frequency of antigen-specific CD4 T cells. Thus intranodal effector cells and recirculating, rapidly responding central-memory-like cells emerged simultaneously from the third day of a primary Th2 response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Immunologic Memory/immunology , Lymph Nodes/immunology , Th2 Cells/immunology , Animals , Animals, Congenic , Antigens, CD/analysis , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cell Movement/immunology , Cell Proliferation , Gene Expression/immunology , Immunization, Secondary , Immunophenotyping , Interleukin-4/genetics , Interleukin-4/metabolism , Kinetics , Leukocyte Common Antigens/genetics , Lymph Nodes/cytology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, CXCR5/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , Th2 Cells/metabolism , Transplantation Chimera/immunology , Vaccination
13.
Mol Ther ; 17(1): 65-72, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19002167

ABSTRACT

Kupffer cells are the resident macrophage population of the liver and have previously been implicated in the pathogenesis of hepatic ischemia-reperfusion injury (IRI). Kupffer cells are the major site of expression of hepatic heme oxygenase-1 (HO-1), which has been shown to have anti-inflammatory actions and to protect animals and cells from oxidative injury. Kupffer cells and circulating monocytes were selectively ablated using liposomal clodronate (LC) in the CD11b DTR mouse before induction of hepatic ischemia. Kupffer cell depletion resulted in loss of HO-1 expression and increased susceptibility to hepatic IRI, whereas ablation of circulating monocytes did not affect IRI phenotype. Targeted deletion of HO-1 rendered mice highly susceptible to hepatic IRI. In vivo, HO-1 deletion resulted in pro-inflammatory Kupffer cell differentiation characterized by enhanced Ly6c and MARCO (macrophage receptor with collagenous structure) expression as well as decreased F4/80 expression, mirrored by an expansion in immature circulating monocytes. In vitro, HO-1 inhibition throughout macrophage differentiation led to increased cell numbers, and pro-inflammatory Ly6c+ CD11c- F4/80- phenotype. These data support a critical role for tissue-resident macrophages in homeostasis following ischemic injury, and a co-dependence of HO-1 expression and tissue-resident macrophage differentiation.


Subject(s)
Heme Oxygenase-1/metabolism , Kupffer Cells/cytology , Kupffer Cells/physiology , Liver/cytology , Reperfusion Injury/immunology , Animals , Antigens, Differentiation/metabolism , Antigens, Ly/metabolism , Blotting, Western , Cell Differentiation , Flow Cytometry , Heme Oxygenase-1/physiology , Immunohistochemistry , Kupffer Cells/metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Receptors, Immunologic/metabolism , Reperfusion Injury/metabolism
14.
Elife ; 92020 05 22.
Article in English | MEDLINE | ID: mdl-32441253

ABSTRACT

Dysregulated cytokine expression by T cells plays a pivotal role in the pathogenesis of autoimmune diseases. However, the identification of the corresponding pathogenic subpopulations is a challenge, since a distinction between physiological variation and a new quality in the expression of protein markers requires combinatorial evaluation. Here, we were able to identify a super-functional follicular helper T cell (Tfh)-like subpopulation in lupus-prone NZBxW mice with our binning approach "pattern recognition of immune cells (PRI)". PRI uncovered a subpopulation of IL-21+ IFN-γhigh PD-1low CD40Lhigh CXCR5- Bcl-6- T cells specifically expanded in diseased mice. In addition, these cells express high levels of TNF-α and IL-2, and provide B cell help for IgG production in an IL-21 and CD40L dependent manner. This super-functional T cell subset might be a superior driver of autoimmune processes due to a polyfunctional and high cytokine expression combined with Tfh-like properties.


Subject(s)
Lupus Erythematosus, Systemic/immunology , Pattern Recognition, Automated/methods , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigens, Differentiation/metabolism , Autoimmunity , B-Lymphocytes/immunology , Coculture Techniques , Female , Flow Cytometry/methods , Immunologic Memory , Interleukins/metabolism , Lymphocyte Activation , Mice , Mice, Inbred NZB
15.
Cell Rep ; 32(5): 107982, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32755576

ABSTRACT

The persistence of long-lived memory plasma cells in the bone marrow depends on survival factors available in the bone marrow, which are provided in niches organized by stromal cells. Using an ex vivo system in which we supply the known survival signals, direct cell contact to stromal cells, and the soluble cytokine a proliferation-inducing ligand (APRIL), we have elucidated the critical signaling pathways required for the survival of long-lived plasma cells. Integrin-mediated contact of bone marrow plasma cells with stromal cells activates the phosphatidylinositol 3-kinase (PI3K) signaling pathway, leading to critical inactivation of Forkhead-Box-Protein O1/3 (FoxO1/3) and preventing the activation of mitochondrial stress-associated effector caspases 3 and 7. Accordingly, inhibition of PI3K signaling in vivo ablates bone marrow plasma cells. APRIL signaling, by the nuclear factor κB (NF-κB) pathway, blocks activation of the endoplasmic-reticulum-stress-associated initiator caspase 12. Thus, stromal-cell-contact-induced PI3K and APRIL-induced NF-κB signaling provide the necessary and complementary signals to maintain bone marrow memory plasma cells.


Subject(s)
Endoplasmic Reticulum Stress , Immunologic Memory , Mitochondria/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Plasma Cells/cytology , Plasma Cells/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , Bone Marrow Cells/metabolism , Caspases/metabolism , Cell Death , Cell Survival , Down-Regulation , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O3/metabolism , Interferon Regulatory Factors/metabolism , Mice, Inbred C57BL , Signal Transduction , Stromal Cells/metabolism
16.
Mol Immunol ; 45(13): 3558-66, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18582945

ABSTRACT

CD4 T helper (Th) cell differentiation defined by in vitro cytokine-directed culture systems leaves major gaps in our knowledge of the mechanisms driving divergent Th differentiation. This is evident from our analysis of the response of mouse ovalbumin-specific CD4 T cells to different forms of ovalbumin that induce markedly distinct responses in vivo. We show that live attenuated ovalbumin-expressing Salmonella (SalOVA) induce Th1-associated T-bet and IFN-gamma. Conversely, alum-precipitated ovalbumin (alumOVA) induces the Th2-associated GATA-3 and IL-4. The early diversity occurring within these CD4 T cells isolated 3 days after immunization was assessed using real-time RT-PCR microfluidic cards designed with 384 selected genes. The technique was validated both at the population and single cell levels at different stages of the responses, showing beta2-microglobulin to be a more stably expressed reference mRNA than either beta-actin or 18S RNA. SalOVA was then shown selectively to induce the OVA-specific CD4 T cells to produce many chemokines and pro-inflammatory cytokines, contrasting with alumOVA-induced cells that only produced a few Th2-associated cytokines. Several cytokines and features associated with follicular helper functions were induced in the OVA-specific CD4 T cells by both antigens. Finally, IL-17RB is strongly associated with OVA-specific CD4 T cells responding to alumOVA, suggesting that alum may promote Th2 immune response through a role for the IL-25/IL-17RB pathway.


Subject(s)
Alum Compounds/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Ovalbumin/immunology , Ovalbumin/metabolism , Salmonella/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression Profiling , Gene Expression Regulation/immunology , Immunization , Immunoprecipitation , Interferon-gamma/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Ovalbumin/drug effects , Salmonella/metabolism , Salmonella Infections/genetics , Salmonella Infections/immunology , Salmonella Infections/metabolism , Salmonella Infections/therapy , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/physiology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/physiology
17.
Front Immunol ; 9: 381, 2018.
Article in English | MEDLINE | ID: mdl-29541075

ABSTRACT

Neonates and infants are more vulnerable to infections and show reduced responses to vaccination. Consequently, repeated immunizations are required to induce protection and early life vaccines against major pathogens such as influenza are yet unavailable. Formulating antigens with potent adjuvants, including immunostimulators and delivery systems, is a demonstrated approach to enhance vaccine efficacy. Yet, adjuvants effective in adults may not meet the specific requirements for activating the early life immune system. Here, we assessed the neonatal adjuvanticity of three novel adjuvants including TLR4 (glucopyranosyl lipid adjuvant-squalene emulsion), TLR9 (IC31®), and Mincle (CAF01) agonists, which all induce germinal centers (GCs) and potent antibody responses to influenza hemagglutinin (HA) in adult mice. In neonates, a single dose of HA formulated into each adjuvant induced T follicular helper (TFH) cells. However, only HA/CAF01 elicited significantly higher and sustained antibody responses, engaging neonatal B cells to differentiate into GCs already after a single dose. Although antibody titers remained lower than in adults, HA-specific responses induced by a single neonatal dose of HA/CAF01 were sufficient to confer protection against influenza viral challenge. Postulating that the neonatal adjuvanticity of CAF01 may result from the functionality of the C-type lectin receptor (CLR) Mincle in early life we asked whether other C-type lectin agonists would show a similar neonatal adjuvanticity. Replacing the Mincle agonist trehalose 6,6'-dibehenate by Curdlan, which binds to Dectin-1, enhanced antibody responses through the induction of similar levels of TFH, GCs and bone marrow high-affinity plasma cells. Thus, specific requirements of early life B cells may already be met after a single vaccine dose using CLR-activating agonists, identified here as promising B cell immunostimulators for early life vaccines when included into cationic liposomes.


Subject(s)
Adjuvants, Immunologic , B-Lymphocytes/immunology , Germinal Center/immunology , Glycolipids/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , beta-Glucans/immunology , Adjuvants, Immunologic/pharmacology , Animals , Animals, Newborn , Antibodies, Viral/blood , Female , Glycolipids/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Lectins, C-Type/agonists , Lectins, C-Type/metabolism , Liposomes , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Toll-Like Receptor 4/agonists , Toll-Like Receptor 9/metabolism , beta-Glucans/pharmacology
18.
Methods Mol Biol ; 1643: 85-103, 2017.
Article in English | MEDLINE | ID: mdl-28667531

ABSTRACT

Immunoglobulins (Igs)-or antibodies (Ab)-are important to combat foreign pathogens but also to the immune system homeostasis. We developed the AID-/-µS-/- mouse model devoid of total soluble Igs and suitable to monitor the role of Igs on immune homeostasis. We used this experimental system to uncover a negative feedback control of marginal zone (MZ) and B1 B cells numbers by naturally secreted Igs. We raised AID-/-µS-/- mice in germ-free conditions demonstrating that this effect of natural secreted Igs is independent of the microbiota. Herein, we provide a comprehensive description of the protocols to establish and use the AID-/-µS-/- mice to study the role of total secreted Igs or of different Ig classes. This study involves Igs injections to AID-/-µS-/- mice or establishment of AID-/-µS-/- mixed bone marrow chimeras that provide a powerful system to study AID-/-µS-/- B cells in the presence of stable concentrations of different Ig classes. While we describe flow cytometric and histological methods to analyze MZ and B1 B cell subsets, AID-/-µS-/- mice can be used to study the effects of natural Igs on other B cell subsets or immune cells.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Immunoglobulins/genetics , Immunoglobulins/immunology , Microbiota/immunology , Animals , Bone Marrow Transplantation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Genotype , Immunoglobulin M/immunology , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Confocal , Transplantation Chimera
19.
Curr Opin Immunol ; 41: 1-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27104290

ABSTRACT

The challenge for any vaccine design is to elicit protective humoral and/or cytotoxic immunity against life threatening pathogens while remaining innocuous. Neonatal vaccinology faces additional challenges linked to intrinsic peculiarities of the innate and adaptive neonatal immune system. These include anti-inflammatory rather than pro-inflammatory responses to innate signals, preferential Th2 differentiation limiting the induction of Th1 and cytotoxic responses, trends to immunoregulatory responses and weak plasma cell and germinal centre B cell responses. Recent progresses in our understanding of the molecular bases of these physiological peculiarities and of the mode of action of novel adjuvants open new opportunities to design vaccine formulations and immunization strategies better adapted to the early life period.


Subject(s)
Aging/immunology , Vaccination , Vaccines/immunology , Adjuvants, Immunologic , Humans
20.
Oncotarget ; 7(27): 41053-41066, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27203391

ABSTRACT

Dendritic cells (DCs) hold promise for anti-cancer immunotherapy. However, clinically, their efficiency is limited and novel strategies to improve DC-mediated anti-tumor responses are needed. Human DCs display high content of sialic acids, which inhibits their maturation and co-stimulation capacity. Here, we aimed to understand whether exogenous desialylation of DCs improves their anti-tumor immunity. Compared to fully sialylated DCs, desialylated human DCs loaded with tumor-antigens showed enhanced ability to induce autologous T cells to proliferate, to secrete Th1 cytokines, and to specifically induce tumor cell apoptosis. Desialylated DCs showed an increased expression of MHC-I and -II, co-stimulatory molecules and an augmented secretion of IL-12. Desialylated HLA-A*02:01 DCs pulsed with gp100 peptides displayed enhanced peptide presentation through MHC-I, resulting in higher activation ofgp100280-288 specific CD8+ cytotoxic T cells. Desialylated murine DCs also exhibited increased MHC and co-stimulatory molecules and higher antigen cross-presentation via MHC-I. These DCs showed higher ability to activate antigen-specific CD4+ and CD8+ T cells, and to specifically induce tumor cell apoptosis. Collectively, our data demonstrates that desialylation improves DCs' ability to elicit T cell-mediated anti-tumor activity, due to increased MHC-I expression and higher antigen presentation via MHC-I. Sialidase treatment of DCs may represent a technology to improve the efficacy of antigen loaded-DC-based vaccines for anti-cancer immunotherapy.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , N-Acetylneuraminic Acid/metabolism , Neoplasms/therapy , Animals , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cells, Cultured , Female , Humans , Immunotherapy/methods , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL