Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Immunol ; 199(12): 4091-4102, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29127145

ABSTRACT

T cell expression of TIM-3 following Ag encounter has been associated with a continuum of functional states ranging from effector memory T cells to exhaustion. We have designed an in vitro culture system to specifically address the impact of anti-TIM-3/TIM-3 engagement on human Ag-specific CD8 T cells during a normal response to Ag and found that anti-TIM-3 treatment enhances T cell function. In our in vitro T cell culture system, MART1-specific CD8 T cells were expanded from healthy donors using artificial APCs. To ensure that the T cells were the only source of TIM-3, cells were rechallenged with peptide-loaded artificial APCs in the presence of anti-TIM-3 Ab. In these conditions, anti-TIM-3 treatment promotes generation of effector T cells as shown by acquisition of an activated phenotype, increased cytokine production, enhanced proliferation, and a transcription program associated with T cell differentiation. Activation of mTORC1 has been previously demonstrated to enhance CD8 T cell effector function and differentiation. Anti-TIM-3 drives CD8 T cell differentiation through activation of the mTORC1 as evidenced by increased levels of phosphorylated S6 protein and rhebl1 transcript. Altogether these findings suggest that anti-TIM-3, together with Ag, drives differentiation in favor of effector T cells via the activation of mTOR pathway. To our knowledge, this is the first report demonstrating that TIM-3 engagement during Ag stimulation directly influences T cell differentiation through mTORC1.


Subject(s)
Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Hepatitis A Virus Cellular Receptor 2/immunology , Immunologic Memory/immunology , Mechanistic Target of Rapamycin Complex 1/immunology , Antibodies, Monoclonal/pharmacology , Cell Division , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation/immunology , Humans , Lymphocyte Activation , Lymphokines/biosynthesis , Lymphokines/genetics , MART-1 Antigen/immunology , Phosphorylation , Protein Processing, Post-Translational , T-Cell Antigen Receptor Specificity , ras Proteins/biosynthesis , ras Proteins/genetics
2.
Front Immunol ; 7: 221, 2016.
Article in English | MEDLINE | ID: mdl-27379090

ABSTRACT

PD-1, TIM-3, and LAG-3 are molecules shown to have immune modulatory properties, and although initially classified as indicators of T cell hyporesponsiveness, it has become clear that they are also associated with the normal course of T cell activation. Functional studies have focused mainly on CD8(+) T cells during chronic inflammation due to interest in co-opting the cellular immune response to eliminate viral or cancerous threats; however, there remains a relative lack of data regarding the expression of these molecules on CD4(+) T cells. Here, we report that expression of the immune checkpoint (IC) molecules PD-1, LAG-3, and TIM-3 are differentially expressed on CD4(+) and CD8(+) T cells in the allogeneic response resulting from a mixed lymphocyte reaction. In these studies, PD-1 expression is higher on CD4(+) T cells compared to CD8(+) T cells. In contrast, TIM-3 is expressed at higher levels on CD8(+) T cells compared to CD4(+) T cells with an apparent reciprocity in that PD-1(+) CD4(+) T cells are frequently TIM-3(lo/-), while TIM-3-expressing CD8(+) T cells are largely PD-1(lo/-). In addition, there is a decrease in the frequency of TIM-3(+) CD4(+) cells producing IFN-γ and IL-5 compared to TIM-3(+) CD8(+) cells. Lastly, the memory T cell phenotype within each IC-expressing subset differs between CD4(+) and CD8(+) T cells. These findings highlight key differences in IC expression patterns between CD4(+) and CD8(+) T cells and may allow for more effective therapeutic targeting of these molecules in the future.

3.
Immun Inflamm Dis ; 3(3): 247-64, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26417440

ABSTRACT

In vitro studies have demonstrated that the immunoreceptor tyrosine-based inhibitory motif (ITIM) of the inhibitory Fc receptor FcγRIIB is critical for mediating attenuation of signaling via immunoreceptor tyrosine-based activation motif (ITAM) containing receptors, such as the B cell antigen receptor (BCR), when FcγRIIB is co-cross-linked to these activation receptors. To test the role of the FcγRIIB ITIM motif in regulation of the B cell immune response in vivo, we constructed lines of transgenic mice expressing a form of FcγRIIB with an inactivating tyrosine (Y) to phenylalanine (F) mutation in the ITIM motif. Detailed studies of one of these lines, in which the mutant FcγRIIB was expressed on B cells and other cell types that normally express this receptor, were performed. No quantitative differences in germinal center (GC) B cell responses were observed between the mutant FcγRIIB transgenic line and control mice. However, serum antibody and antibody forming cell responses were often observed to be elevated in the ITIM mutant FcγRIIB transgenic mice as compared to controls, though not to the same extent as mice deficient in expression of FcγRIIB. Moreover, primary B cells from the ITIM mutant FcγRIIB line did not display the same level of augmented BCR signaling as primary FcγRIIB deficient B cells under conditions inducing co-cross-linking of FcγRIIB and the BCR. In total, these data suggest that a functional ITIM motif is not required for all in vivo inhibitory activity of this receptor. However, we also found that the transgenic ITIM mutant FcγRIIB receptor was expressed at abnormal levels in several hematopoietic lineages. Thus, confirmation of our findings will require the generation and analysis of mice in which an ITIM mutant form of FcγRIIB is expressed in vivo as is the endogenous receptor.

4.
AIDS Res Hum Retroviruses ; 31(11): 1126-38, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25848984

ABSTRACT

We previously showed that a matrix (M) gene-deleted rabies virus (RABV)-based vaccine (RABV-ΔM) is highly immunogenic and induces potent B cell responses in the context of RABV infection. We speculated that RABV-ΔM expressing HIV proteins would also induce potent B cell responses against HIV antigens. As a prerequisite to future studies in nonhuman primates, we completed immunogenicity studies in mice to confirm the ability of RABV-ΔM to induce polyfunctional B cell responses in the context of HIV. To that end, the envelope protein from the mac239 strain of SIV (SIVmac239Env) was cloned into RABV-ΔM, resulting in RABV-ΔM-Env. Infectious virus was recovered following standard methods and propagated on baby hamster kidney cells stably expressing RABV M [>10(7) focus forming units (ffu)/ml]. Western blot analysis of cell lysates or of purified virions confirmed Env expression on the surface of infected cells and within virus particles, respectively. Positive neutralization activity against a neutralization-sensitive SIV strain and to a lesser extent against a neutralization-resistant SIV strain was detected in mice after a single intramuscular inoculation with RABV-ΔM-Env. The quality, but not quantity, of the antibody response was enhanced via boosting with recombinant gp130 or RABV-ΔM-Env as measured by an increase in antibody avidity and a skewing toward a Th1-type antibody response. We also show that an intradermal inoculation induces higher antibodies than an intramuscular or intranasal inoculation. An intradermal inoculation of RABV-ΔM-Env followed by a boost inoculation with recombinant gp130 produced anti-SIV antibodies with neutralizing and nonneutralizing antibody (nNAb) effector functions. Together, RABV-ΔM-Env induces B cells to secrete antibodies against SIV with the potential to clear both "free" and cell-associated virus. Strategies capable of eliciting both NAbs as well as nNAbs might help to improve the efficacy of HIV-1 vaccines.


Subject(s)
Antibodies, Viral/blood , Antibody Formation , Rabies Vaccines/immunology , Rabies virus/immunology , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , Animals , B-Lymphocytes/immunology , Female , Injections, Intradermal , Injections, Intramuscular , Mice, Inbred C57BL , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Rabies virus/genetics , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/genetics , Simian Immunodeficiency Virus/genetics
5.
J Virol ; 89(5): 2842-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25540370

ABSTRACT

UNLABELLED: Replication-deficient rabies virus (RABV)-based vaccines induce rapid and potent antibody responses via T cell-independent and T cell-dependent mechanisms. To further investigate early events in vaccine-induced antibody responses against RABV infections, we studied the role of macrophages as mediators of RABV-based vaccine immunogenicity. In this report, we show that a recombinant matrix gene-deleted RABV-based vaccine (rRABV-ΔM) infects and activates primary murine macrophages in vitro. Immunization of mice with live RABV-based vaccines results in accumulation of macrophages at the site of immunization, which suggests that macrophages in tissues support the development of effective anti-RABV B cell responses. However, we show that draining lymph node macrophages, but not macrophages at the site of immunization, are essential for the generation of germinal center B cells, follicular T helper cells, and RABV-specific antibodies. Our findings have implications for the design of new RABV-based vaccines for which early immunological events are important for the protection against RABV in postexposure settings. IMPORTANCE: More than two-thirds of the world's population live in regions where rabies is endemic. Postexposure prophylaxis is the primary means of treating humans. Identifying immunological principles that guide the development of rapid and potent antibody responses against rabies infections will greatly increase our ability to produce more-effective rabies vaccines. Here we report that macrophages in the draining lymph node, but not in the tissue at the site of immunization are important for vaccine-induced antibody responses to rabies. Information gleaned from this study may help guide the development of a single-dose vaccine against rabies infections.


Subject(s)
Antibody Formation , B-Lymphocytes/immunology , Germinal Center/immunology , Lymph Nodes/immunology , Macrophages/immunology , Rabies Vaccines/immunology , Animals , Antibodies, Viral/blood , Female , Injections, Intradermal , Mice, Inbred C57BL , Rabies Vaccines/administration & dosage , T-Lymphocytes, Helper-Inducer/immunology
6.
PLoS One ; 9(1): e87098, 2014.
Article in English | MEDLINE | ID: mdl-24489846

ABSTRACT

We have previously shown that live-attenuated rabies virus (RABV)-based vaccines infect and directly activate murine and human primary B cells in-vitro, which we propose can be exploited to help develop a single-dose RABV-based vaccine. Here we report on a novel approach to utilize the binding of Intracellular Adhesion Molecule-1 (ICAM-1) to its binding partner, Lymphocyte Function-associated Antigen-1 (LFA-1), on B cells to enhance B cell activation and RABV-specific antibody responses. We used a reverse genetics approach to clone, recover, and characterize a live-attenuated recombinant RABV-based vaccine expressing the murine Icam1 gene (rRABV-mICAM-1). We show that the murine ICAM-1 gene product is incorporated into virus particles, potentially exposing ICAM-1 to extracellular binding partners. While rRABV-mICAM-1 showed 10-100-fold decrease in viral titers on baby hamster kidney cells compared to the parental virus (rRABV), rRABV-mICAM-1 infected and activated primary murine B cells in-vitro more efficiently than rRABV, as indicated by significant upregulation of CD69, CD40, and MHCII on the surface of infected B cells. ICAM-1 expression on the virus surface was responsible for enhanced B cell infection since pre-treating rRABV-mICAM-1 with a neutralizing anti-ICAM-1 antibody reduced B cell infection to levels observed with rRABV alone. Furthermore, 100-fold less rRABV-mICAM-1 was needed to induce antibody titers in immunized mice equivalent to antibody titers observed in rRABV-immunized mice. Of note, only 10(3) focus forming units (ffu)/mouse of rRABV-mICAM-1 was needed to induce significant anti-RABV antibody titers as early as five days post-immunization. As both speed and potency of antibody responses are important in controlling human RABV infection in a post-exposure setting, these data show that expression of Icam1 from the RABV genome, which is then incorporated into the virus particle, is a promising strategy for the development of a single-dose RABV vaccine that requires only a minimum of virus.


Subject(s)
Antibodies, Viral/biosynthesis , Genome, Viral , Intercellular Adhesion Molecule-1/immunology , Rabies Vaccines/immunology , Rabies virus/immunology , Rabies/prevention & control , Viral Proteins/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/virology , Cell Line , Cricetinae , Female , Gene Expression , Humans , Immunity, Humoral/drug effects , Intercellular Adhesion Molecule-1/genetics , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mice, Inbred C57BL , Rabies/immunology , Rabies/virology , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Rabies virus/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccines, Attenuated , Vaccines, Synthetic , Viral Proteins/genetics , Virion/genetics , Virion/immunology
7.
J Virol ; 87(16): 9097-110, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23760241

ABSTRACT

Replication-deficient rabies viruses (RABV) are promising rabies postexposure vaccines due to their prompt and potent stimulation of protective virus neutralizing antibody titers, which are produced in mice by both T-dependent and T-independent mechanisms. To promote such early and robust B cell stimulation, we hypothesized that live RABV-based vaccines directly infect B cells, thereby activating a large pool of antigen-presenting cells (APCs) capable of providing early priming and costimulation to CD4(+) T cells. In this report, we show that live RABV-based vaccine vectors efficiently infect naive primary murine and human B cells ex vivo. Infection of B cells resulted in the significant upregulation of early markers of B cell activation and antigen presentation, including CD69, major histocompatibility complex class II (MHC-II), and CD40 in murine B cells or HLA-DR and CD40 in human B cells compared to mock-infected cells or cells treated with an inactivated RABV-based vaccine. Furthermore, primary B cells infected with a live RABV expressing ovalbumin were able to prime and stimulate naive CD4(+) OT-II T cells to proliferate and to secrete interleukin-2 (IL-2), demonstrating a functional consequence of B cell infection and activation by live RABV-based vaccine vectors. We propose that this direct B cell stimulation by live RABV-based vaccines is a potential mechanism underlying their induction of early protective T cell-dependent B cell responses, and that designing live RABV-based vaccines to infect and activate B cells represents a promising strategy to develop a single-dose postexposure rabies vaccine where the generation of early protective antibody titers is critical.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/virology , Lymphocyte Activation , Rabies Vaccines/immunology , Rabies virus/immunology , Adult , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cells, Cultured , Female , Humans , Interleukin-2/metabolism , Male , Mice , Mice, Inbred C57BL , Vaccines, Attenuated/immunology
8.
J Virol ; 87(16): 9217-22, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23760250

ABSTRACT

B cells secreting IgG antibodies, but not IgM, are thought to be solely responsible for vaccine-induced protection against rabies virus (RABV) infections in postexposure settings. In this report, we reinvestigated the potential for IgM to mediate protection in a mouse model of RABV vaccination. Immunocompetent mice immunized with an experimental live replication-deficient RABV-based vaccine produced virus neutralizing antibodies (VNAs) within 3 days of vaccination. However, mice unable to produce soluble IgM (sIgM(-/-)) did not produce VNAs until 7 days postimmunization. Furthermore, sIgM(-/-) mice were not protected against RABV infection when challenged 3 days postimmunization, while all wild-type mice survived challenge. Consistent with the lack of protection against pathogenic RABV challenge, approximately 50- to 100-fold higher viral loads of challenge virus were detected in the muscle, spinal cord, and brain of immunized sIgM(-/-) mice compared to control mice. In addition, IgG antibody titers in vaccinated wild-type and sIgM(-/-) mice were similar at all time points postimmunization, suggesting that protection against RABV challenge is due to the direct effects of IgM and not the influence of IgM on the development of effective IgG antibody titers. In all, early vaccine-induced IgM can limit dissemination of pathogenic RABV to the central nervous system and mediate protection against pathogenic RABV challenge. Considering the importance for the rapid induction of VNAs to protect against RABV infections in postexposure prophylaxis settings, these findings may help guide the development of a single-dose human rabies vaccine.


Subject(s)
Antibodies, Viral/immunology , Immunoglobulin M/immunology , Post-Exposure Prophylaxis/methods , Rabies Vaccines/immunology , Rabies virus/immunology , Rabies/prevention & control , Vaccination/methods , Animal Structures/virology , Animals , Disease Models, Animal , Female , Immunoglobulin M/deficiency , Mice , Rabies/immunology , Rabies Vaccines/administration & dosage , Survival Analysis , Viral Load
9.
J Immunol ; 190(10): 4923-7, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23567932

ABSTRACT

We demonstrated recently that, after accumulation of Ag-engaged B cells at the T cell zone boundaries in the spleen, these B cells migrate to the perimeter of follicles adjacent to the marginal zone. They undergo rapid proliferation at this site prior to coalescence into germinal centers (GCs). In this article, we report that this phase of migration and expansion of activated Ag-specific B cells, as well as subsequent formation of GCs, does not take place in the absence of splenic macrophages. Our data suggest a previously unappreciated function for macrophages in orchestrating the early phases of T cell-dependent B cell responses and formation of GCs distinct from their potential role in Ag presentation to T cells.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Lymphocyte Activation/immunology , Macrophages/immunology , Adoptive Transfer , Animals , Cell Movement , Cell Proliferation , Clodronic Acid/pharmacology , Liposomes/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/immunology , T-Lymphocytes/immunology
10.
J Immunol ; 188(1): 47-56, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22131331

ABSTRACT

Secondary Ab V region gene segment rearrangement, termed receptor editing, is a major mechanism contributing to B lymphocyte self-tolerance. However, the parameters that determine whether a B cell undergoes editing are a current subject of debate. We tested the role that the level of BCR expression plays in the regulation of receptor editing in a polyclonal population of B cells differentiating in vivo. Expression of a short hairpin RNA for κ L chain RNA in B cells resulted in reduction in levels of this RNA and surface BCRs. Strikingly, fully mature and functional B cells that developed in vivo and efficiently expressed the short hairpin RNA predominantly expressed BCRs containing λ light chains. This shift in L chain repertoire was accompanied by inhibition of development, increased Rag gene expression, and increased λ V gene segment-cleavage events at the immature B cell stage. These data demonstrated that reducing the translation of BCRs that are members of the natural repertoire at the immature B cell stage is sufficient to promote editing.


Subject(s)
Gene Expression Regulation/immunology , Immunoglobulin kappa-Chains/immunology , Immunoglobulin lambda-Chains/immunology , RNA Editing/immunology , RNA, Messenger/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Cell Line , Gene Expression Regulation/genetics , Immunoglobulin kappa-Chains/biosynthesis , Immunoglobulin kappa-Chains/genetics , Immunoglobulin lambda-Chains/biosynthesis , Immunoglobulin lambda-Chains/genetics , Mice , RNA Editing/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Antigen, B-Cell/biosynthesis , Receptors, Antigen, B-Cell/genetics
11.
J Immunol ; 182(1): 398-407, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109171

ABSTRACT

We have described an Ig-transgenic, autoreactive B cell clonotype that undergoes a novel tolerance pathway. Early in development this clonotype expresses average BCR levels, but these levels are progressively down-regulated as development proceeds efficiently to the mature, follicular compartment. This clonotype does not display conventional features of anergy and can be induced to undergo apoptosis and receptor editing in in vitro bone marrow cultures, but these pathways are not taken in vivo. These data suggested that autoantigen-driven down-regulation of BCR levels and, hence, avidity for autoantigen allows this clonotype to bypass conventional tolerance mechanisms. To test this idea, we enforced elevated levels of expression of BCR in this clonotype by making the transgenic Igh locus homozygous. This resulted in retarded clonotype development and L chain receptor editing in vivo. These data support a pivotal role for adaptive, autoantigen-induced adjustment of BCR expression levels in the regulation of primary B cell development and tolerance.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Immune Tolerance , Receptors, Antigen, B-Cell/biosynthesis , Receptors, Antigen, B-Cell/genetics , Signal Transduction/immunology , Animals , Antibodies, Antinuclear/physiology , Autoantigens/metabolism , Autoantigens/physiology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cells, Cultured , Down-Regulation/genetics , Down-Regulation/immunology , Growth Inhibitors/metabolism , Growth Inhibitors/physiology , Immune Tolerance/genetics , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA Editing/genetics , RNA Editing/immunology , Receptors, Antigen, B-Cell/antagonists & inhibitors , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/genetics
12.
Immunogenetics ; 56(10): 743-53, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15578175

ABSTRACT

Toll-like receptors (TLRs) play an important role in the recognition of microbial components. Only chicken TLR2 and -4 have been reported in the literature. The objectives of this study were to identify new chicken TLRs and to evaluate evolutionary significance of these receptors. Searching chicken genomic databases and DNA sequencing revealed five new TLRs, TLR1 (type 1 and 2), -3, -5, and -7. No chicken orthologues of mammalian TLR8, -9, or -10 were found. As in mammals, all chicken TLRs (chTLRs) share identical protein secondary structure that consists of several leucine-rich domains, a transmembrane domain, and Toll/Interleukin-1 receptor domain(s). Phylogenetic analyses indicate that the identified chTLR genes are the orthologues of TLRs in mammals. Analyses of the number of synonymous substitutions per synonymous site and nonsynonymous substitutions per nonsynonymous site indicate that the nucleotide sequences coding for the leucine-rich repeats of chicken TLR1 type 1 and type 2 were significantly under positive Darwinian selection. In contrast, the sequences of other TLRs were under purifying selection. These results support the hypothesis that one of the major evolutionary strategies of the innate immune system is to recognize a few highly conserved microbial components with several conserved TLRs. The results also indicate that the sequence changes in the ligand-binding domains of TLR1 in chickens provide adaptive advantages during evolution.


Subject(s)
Chickens/immunology , Membrane Glycoproteins/analysis , Receptors, Cell Surface/analysis , Amino Acid Sequence , Animals , Chromosome Mapping , Gene Expression Profiling , Leucine-Rich Repeat Proteins , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Molecular Sequence Data , Phylogeny , Proteins/analysis , Proteins/chemistry , Proteins/genetics , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Toll-Like Receptor 1 , Toll-Like Receptor 2 , Toll-Like Receptor 8 , Toll-Like Receptors
SELECTION OF CITATIONS
SEARCH DETAIL
...