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










Publication year range
1.
Pest Manag Sci ; 72(10): 1965-76, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26818964

ABSTRACT

BACKGROUND: Biobeds are used for on-farm bioremediation of pesticides in sprayer rinsate and from spills during sprayer filling. Using locally sourced materials from Saskatchewan, Canada, a biobed matrix was evaluated for its effectiveness for mineralising and degrading 2,4-dichlorophenoxyacetic acid dimethylamine salt (2,4-D DMA) compared with the topsoil used in the biobed matrix. RESULTS: Applying 2,4-D DMA to the biobed matrix caused a 2-3 day lag in CO2 production not observed when the herbicide was applied to topsoil. Despite the initial lag, less residual 2,4-D was measured in the biobed (0%) matrix than in the topsoil (57%) after a 28 day incubation. When the herbicide was applied 5 times to the biobed matrix, net CO2 increased immediately after each 2,4-D DMA application. Mineralisation of 2,4-D DMA was 61.9% and residual 2,4-D in the biobed matrix was 0.3% after 60 days, compared with corresponding values of 32.9 and 70.9% in topsoil. CONCLUSION: The biobed matrix enhanced the mineralisation and degradation of 2,4-D DMA, indicating the potential for successful implementation of biobeds under Canadian conditions. The biobed matrix was more effective for mineralising and degrading the herbicide compared with the topsoil used in the biobed matrix. By correcting for biobed matrix and formulation blank, CO2 evolution was a reliable indicator of 2,4-D DMA mineralisation. © 2016 Society of Chemical Industry.


Subject(s)
2,4-Dichlorophenoxyacetic Acid/metabolism , Dimethylamines/metabolism , Herbicides/metabolism , Soil/chemistry , Biodegradation, Environmental , Carbon Dioxide/chemistry , Environmental Pollution/prevention & control , Saskatchewan
2.
J Mol Biol ; 399(1): 113-9, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20382165

ABSTRACT

Adiponectin is an adipocyte-derived hormone that has been shown to play important roles in the regulation of glucose and energy homeostasis. It exists as homotrimers or complexes containing multiple homotrimer units in plasma. The recombinant adiponectin proteins have been difficult to produce, making it challenging for both research as well as potential therapeutic development. Here, we show a novel approach for the generation of globular adiponectin that involves linking three monomer sequences together in tandem to generate one continuous polypeptide, which we have termed single-chain globular adiponectin (sc-gAd). To improve the pharmacokinetic properties of sc-gAd further, we fused it to an Fc fragment. The combined effects of single-chain and Fc fusion improved the plasma half-life from less than 2 h to close to 2 weeks. Using adeno-associated virus as a delivery method, we demonstrate that Fc-sc-gAd improved both fasting glucose levels and the tolerance to a glucose challenge in ob/ob mice without changes in body weight. Therefore, our results demonstrated the feasibility of generating globular adiponectin trimers from a single polypeptide and a long-acting globular adiponectin that could serve as a starting point for adiponectin-based therapeutics. This novel approach could also be applied to other complement factor C1q family members; in particular, this opens the possibility to study the biological functions of precisely defined heterotrimers of various family members that had not been previously possible.


Subject(s)
Adiponectin/chemistry , Recombinant Proteins/chemistry , Adipocytes/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Animals , Humans , Mice , Mice, Inbred Strains , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
4.
J Immunol ; 182(4): 2213-20, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19201875

ABSTRACT

Exosomes are small membrane vesicles of endocytic origin that are secreted by most cells in culture, but are also present in serum. They contain a wide array of protein ligands on their surface, which has led to the hypothesis that they might mediate intercellular communication. Indeed, data support that exosomes can transfer Ags to dendritic cells (DC), and, interestingly, that these DC can subsequently induce T cell priming or tolerance. We have investigated whether this concept can be expanded to antiviral immunity. We isolated exosomes from supernatant of cultured bone marrow-derived DC (BMDC) that were infected with lymphocytic choriomeningitis virus (LCMV) or loaded with an immunodominant LCMV peptide, and characterized them by flow cytometry upon binding to beads. We then incubated the exosome preparations with BMDC and looked at their potential to activate LCMV gp33-specific naive and memory CD8 T cells. We found that exosomes do not significantly contribute to CD8 T cell cross-priming in vitro. Additionally, exosomes derived from in vitro-infected BMDC did not exhibit significant in vivo priming activity, as evidenced by the lack of protection following exosome vaccination. Thus, DC-derived exosomes do not appear to contribute significantly to CTL priming during acute LCMV infection.


Subject(s)
Cross-Priming/immunology , Dendritic Cells/immunology , Exosomes/immunology , Lymphocytic Choriomeningitis/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Bone Marrow Cells/immunology , Dendritic Cells/virology , Exosomes/virology , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Cytotoxic/virology
5.
J Immunol ; 181(3): 2124-33, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18641351

ABSTRACT

The primary CD8(+) T cell response of C57BL/6J mice against the 28 known epitopes of lymphocytic choriomeningitis virus (LCMV) is associated with a clear immunodominance hierarchy whose mechanism has yet to be defined. To evaluate the role of epitope competition in immunodominance, we manipulated the number of CD8(+) T cell epitopes that could be recognized during LCMV infection. Decreasing epitope numbers, using a viral variant lacking dominant epitopes or C57BL/6J mice lacking H-2K(b), resulted in minor response increases for the remaining epitopes and no new epitopes being recognized. Increasing epitope numbers by using F(1) hybrid mice, delivery by recombinant vaccinia virus, or epitope delivery as a pool in IFA maintained the overall response pattern; however, changes in the hierarchy did become apparent. MHC binding affinity of these epitopes was measured and was found to not strictly predict the hierarchy since in several cases similarly high binding affinities were associated with differences in immunodominance. In these instances the naive CD8(+) T cell precursor frequency, directly measured by tetramer staining, correlated with the response hierarchy seen after LCMV infection. Finally, we investigated an escape mutant of the dominant GP33-41 epitope that elicited a weak response following LCMV variant virus infection. Strikingly, dominance loss likely reflects a substantial reduction in frequencies of naive precursors specific for this epitope. Thus, our results indicate that an intrinsic property of the epitope (MHC binding affinity) and an intrinsic property of the host (naive precursor frequency) jointly dictate the immunodominance hierarchy of CD8(+) T cell responses.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Shape/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens/immunology , Immunity, Innate/immunology , Animals , Histocompatibility Antigens/genetics , Histocompatibility Antigens/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Knockout , Protein Binding
6.
Diabetes ; 56(4): 1059-68, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17395746

ABSTRACT

During an autoimmune process, the autoaggressive response spreads from the initiating autoantigen to other antigens expressed in the target organ. Based on evidence from experimental models for multiple sclerosis, such "antigenic spreading" can play an important role in the exacerbation of clinical disease. We evaluated whether pathogenesis of spontaneous diabetes in NOD mice could be accelerated in a similar way when a novel autoantigen was expressed in pancreatic beta-cells. Unexpectedly, we found that the expression of the lymphocytic choriomeningitis virus nucleoprotein only led to marginal enhancement of diabetes, although such NOD-nucleoprotein mice were not tolerant to nucleoprotein. Although the frequency of nucleoprotein-specific CD8 T-cells in the pancreatic draining lymph node was comparable with the frequency of islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)-specific T-cells, more IGRP-specific CD8 T-cells were found both systemically and in the islets where there was a fourfold increase. Interestingly, and in contrast to nucleoprotein-specific CD8 T-cells, IGRP-specific T-cells showed increased CXCR3 expression. Thus, autoreactivity toward de novo-expressed beta-cell autoantigens will not accelerate autoimmunity unless large numbers of antigen-experienced autoreactive T-cells expressing the appropriate chemokine receptors are present.


Subject(s)
Autoantigens/immunology , Diabetes Mellitus, Type 1/physiopathology , Insulin-Secreting Cells/immunology , Animals , Cloning, Molecular , DNA Primers , Diabetes Mellitus, Type 1/immunology , Enzyme-Linked Immunosorbent Assay , Insulin/analysis , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Inbred NOD , Mice, Transgenic , Nucleoproteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Viral Proteins/genetics
7.
Am J Pathol ; 170(2): 557-66, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17255324

ABSTRACT

We examine here the outcome of viral encephalomyelitis [mouse hepatitis virus (MHV) A59, Theiler's encephalomyelitis virus, and Coxsackievirus B3] in mice with autoantibodies to a central nervous system (CNS)-specific antigen, myelin oligodendrocyte glycoprotein, that usually develop no clinical disease. Morbidity and mortality of the acute viral CNS disease was augmented by the presence of the autoantibodies in all three viral infections. Transfer of serum containing the autoantibodies at the time of infection with MHV was sufficient to reproduce the exacerbated disease. The presence of the autoantibodies was found to result in increased infiltration of mononuclear cells into the brain. Early demyelination was severely augmented in brains and spinal cords of MHV-infected mice with CNS-specific autoantibodies. The antibody-mediated exacerbation was shown to be independent of the complement system but to require expression of Fc receptors, because it was observed in C'-3-deficient but not in Fc receptor-deficient mice. Our study illustrates the possibility that infections can lead to much more profound immunopathology in the presence of an otherwise latent autoimmune condition.


Subject(s)
Autoimmune Diseases/immunology , Brain/immunology , Encephalitis, Viral/immunology , Myelin-Associated Glycoprotein/immunology , Spinal Cord/immunology , Acute Disease , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Autoimmune Diseases/virology , Brain/pathology , Brain/virology , Complement C3/deficiency , Complement C3/immunology , Encephalitis, Viral/genetics , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Mice , Mice, Knockout , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , Receptors, Fc/deficiency , Receptors, Fc/immunology , Spinal Cord/pathology , Spinal Cord/virology , Transgenes/immunology
9.
J Autoimmun ; 27(3): 166-73, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17045460

ABSTRACT

CD8(+) cytotoxic T lymphocytes (CTL) can rapidly kill beta-cells and therefore contribute to the development of type 1 diabetes (T1D). CTL-mediated beta-cell killing can occur via perforin-mediated lysis, Fas-Fas-L interaction, and the secretion of TNF-alpha or IFN-gamma. The secretion of IFN-gamma can contribute to beta-cell death directly by eliciting nitric oxide production, and indirectly by upregulating MHC class I and 'unmasking' beta-cells for recognition by CTL. Earlier studies in the RIP-LCMV mouse model of diabetes showed that disruption of beta-cell IFN-gamma signaling alone abolished the direct detrimental effects of IFN-gamma, but not MHC class I upregulation. Suppressor of cytokine signaling-1 (SOCS-1) represses several crucial cytokine signaling pathways simultaneously, among them IFN-gamma and IL-1-beta. We therefore evaluated the protective capacity of islet cell SOCS-1 expression in the CD8(+) mediated RIP-LCMV diabetes model. Clinical disease was prevented in over 90% of the mice. Not only absence of MHC-I and Fas upregulation, but also resistance to cytokine-induced killing of beta-cells and a complete lack of CXCL-10 (IP10) production in islets led to a lack of islet infiltration and impaired activation of autoaggressive CD4(+) and CD8(+) T-cells in these mice. Thus, SOCS expression renders beta-cells resistant to CTL attack in a mouse model of T1D.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/genetics , Insulin-Secreting Cells/metabolism , Suppressor of Cytokine Signaling Proteins/biosynthesis , Animals , Cytotoxicity, Immunologic , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/virology , Disease Models, Animal , Flow Cytometry , Histocompatibility Antigens Class I/biosynthesis , Immunohistochemistry , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Lymphocytic choriomeningitis virus , Mice , Mice, Transgenic , Microscopy, Confocal , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Cytokine/biosynthesis , Signal Transduction/physiology , Suppressor of Cytokine Signaling 1 Protein , fas Receptor/biosynthesis
10.
J Virol ; 80(1): 270-80, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16352552

ABSTRACT

CTLA-4 is considered one of the most potent negative regulators of T-cell activation. To circumvent experimental limitations due to fatal lymphoproliferative disease associated with genetic ablation of CTLA-4, we have used radiation chimeras reconstituted with a mixture of CTLA-4+/+ and CTLA-4-/- bone marrow that retain a normal phenotype and allow the evaluation of long-term T-cell immunity under conditions of intrinsic CTLA-4 deficiency. Following virus infection, we profiled primary, memory, and secondary CD8+ and CD4+ T-cell responses directed against eight different viral epitopes. Our data demonstrate unaltered antigen-driven proliferation, acquisition of effector functions, distribution of epitope hierarchies, T-cell receptor repertoire selection, functional avidities, and long-term memory maintenance in the absence of CTLA-4. Moreover, regulation of memory T-cell survival and homeostatic proliferation, as well as secondary responses, was equivalent in virus-specific CTLA4+/+ and CTL-A-4-/- T-cell populations. Thus, lack of CTLA-4 expression by antigen-specific T cells can be compensated for by extrinsic factors in the presence of CTLA-4 expression by other cells. These findings have implications for the physiologic, pathological, and therapeutic regulation of costimulation.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , CTLA-4 Antigen , Gene Expression Regulation/immunology , Gene Silencing , Lymphocyte Activation/immunology , Mice , T-Lymphocytes, Regulatory/immunology
11.
Blood ; 106(13): 4225-33, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16118326

ABSTRACT

In immunotherapy, dendritic cells (DCs) can be used as powerful antigen-presenting cells to enhance or suppress antigen-specific immunity upon in vivo transfer in mice or humans. However, to generate sufficient numbers of DCs, most protocols include an ex vivo culture step, wherein the cells are exposed to heterologous serum and/or antigenic stimuli. In mouse models of virus infection and virus-induced autoimmunity, we tested how heterologous serum affects the immunomodulatory capacity of immature DCs generated in the presence of IL-10 by comparing fetal bovine serum (FBS)- or normal mouse serum (NMS)-supplemented DC cultures. We show that FBS-exposed DCs induce a systemic immune deviation characterized by reduction of virus-specific T cells, delayed viral clearance, and enhanced systemic production of interleukin 4 (IL-4), IL-5, and IL-10 to FBS-derived antigens, including bovine serum albumin (BSA). By contrast, DCs generated in NMS-supplemented cultures modulated immunity and autoimmunity in an antigen-specific fashion. These cells did not induce systemic IL-4, IL-5, or IL-10 production and inhibited generation of virus-specific T cells or autoimmunity only if pulsed with a viral antigen. These data underscore the importance of using autologous serum-derived immature DCs in preclinical animal studies to accurately assess their immunomodulatory potential in future human therapeutic settings, where application of FBS is not feasible.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance/immunology , Serum/immunology , Animals , Cattle , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/drug effects , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/immunology , Immune Tolerance/drug effects , Lymphocyte Activation/drug effects , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Mice , Mice, Knockout , Phenotype , T-Lymphocytes/immunology , Th2 Cells/immunology
12.
J Clin Invest ; 114(9): 1290-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15520861

ABSTRACT

We document here that infection of prediabetic mice with a virus expressing an H-2Kb-restricted mimic ligand to a self epitope present on beta cells accelerates the development of autoimmune diabetes. Immunization with the mimic ligand expanded autoreactive T cell populations, which was followed by their trafficking to the islets, as visualized in situ by tetramer staining. In contrast, the mimic ligand did not generate sufficient autoreactive T cells in naive mice to initiate disease. Diabetes acceleration did not occur in H-2Kb-deficient mice or in mice tolerized to the mimic ligand. Thus, arenavirus-expressed mimics of self antigens accelerate a previously established autoimmune process. Sequential heterologous viral infections might therefore act in concert to precipitate clinical autoimmune disease, even if single exposure to a viral mimic does not always cause sufficient tissue destruction.


Subject(s)
Autoantigens/chemistry , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Animals , B-Lymphocytes/immunology , Blood Glucose/metabolism , CD8-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Disease Models, Animal , Epitopes/chemistry , Immunohistochemistry , Ligands , Mice , Mice, Inbred C57BL , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Time Factors , Transgenes
13.
Diabetes ; 53(3): 591-6, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14988242

ABSTRACT

Tissue-specific expression of Fas-ligand (Fas-L) can provide immune privilege by inducing apoptosis of "invading" lymphocytes expressing Fas. However, accelerated diabetes has been reported in transgenic mice expressing Fas-L in islets (RIP-Fas-L) as a result of Fas-dependent fratricide of beta-cells after transfer of diabetogenic clones. Here we studied whether Fas-L could protect islets from autoaggressive CD8 lymphocytes in a transgenic model of virally induced diabetes (RIP-LCMV-NP transgenic mice), in which the autoaggressive response is directed to a viral nucleoprotein (NP) expressed as a transgene in beta-cells. Indeed, disease incidence after viral (lymphocytic choriomeningitis virus [LCMV]) infection was reduced by approximately 30%, which was associated with a decrease of autoaggressive CD8 NP-specific lymphocytes in islets and pancreatic draining lymph nodes. However, surprisingly, a high degree (50%) of diabetes was seen in mice that expressed only Fas-L but not the viral transgene (NP) in beta-cells after infection with LCMV. This was due to induction of Fas on beta-cells after LCMV infection of the pancreas, resulting in Fas/Fas-L-mediated fratricide. Thus, although Fas-L can lend some immune privilege to islet cells, local virus-induced inflammation will induce Fas on beta-cells, leading to their mutual destruction if Fas-L is present. Expression of Fas-L therefore might not be protective in situations in which viral inflammation can be expected, resulting in Fas induction on the targeted cell itself.


Subject(s)
Islets of Langerhans/pathology , Lymphocytic Choriomeningitis/physiopathology , Membrane Glycoproteins/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Crosses, Genetic , Fas Ligand Protein , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Islets of Langerhans/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus , Mice , Mice, Transgenic , Spleen/immunology
14.
J Clin Invest ; 113(1): 74-84, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14702111

ABSTRACT

Viruses can cause but can also prevent autoimmune disease. This dualism has certainly hampered attempts to establish a causal relationship between viral infections and type 1 diabetes (T1D). To develop a better mechanistic understanding of how viruses can influence the development of autoimmune disease, we exposed prediabetic mice to various viral infections. We used the well-established NOD and transgenic RIP-LCMV models of autoimmune diabetes. In both cases, infection with the lymphocytic choriomeningitis virus (LCMV) completely abrogated the diabetic process. Interestingly, such therapeutic viral infections resulted in a rapid recruitment of T lymphocytes from the islet infiltrate to the pancreatic draining lymph node, where increased apoptosis was occurring. In both models this was associated with a selective and extensive expression of the chemokine IP-10 (CXCL10), which predominantly attracts activated T lymphocytes, in the pancreatic draining lymph node, and in RIP-LCMV mice it depended on the viral antigenic load. In RIP-LCMV mice, blockade of TNF-alpha or IFN-gamma in vivo abolished the prevention of T1D. Thus, virally induced proinflammatory cytokines and chemokines can influence the ongoing autoaggressive process beneficially at the preclinical stage, if produced at the correct location, time, and levels.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic choriomeningitis virus/physiology , Prediabetic State/prevention & control , Aging , Animals , Diabetes Mellitus, Type 1/virology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Inbred NOD , Mice, Transgenic , Polymerase Chain Reaction , Prediabetic State/virology , Time Factors
15.
Nature ; 421(6925): 852-6, 2003 Feb 20.
Article in English | MEDLINE | ID: mdl-12594515

ABSTRACT

A long-standing paradox in cellular immunology concerns the conditional requirement for CD4+ T-helper (T(H)) cells in the priming of cytotoxic CD8+ T lymphocyte (CTL) responses in vivo. Whereas CTL responses against certain viruses can be primed in the absence of CD4+ T cells, others, such as those mediated through 'cross-priming' by host antigen-presenting cells, are dependent on T(H) cells. A clearer understanding of the contribution of T(H) cells to CTL development has been hampered by the fact that most T(H)-independent responses have been demonstrated ex vivo as primary cytotoxic effectors, whereas T(H)-dependent responses generally require secondary in vitro re-stimulation for their detection. Here, we have monitored the primary and secondary responses of T(H)-dependent and T(H)-independent CTLs and find in both cases that CD4+ T cells are dispensable for primary expansion of CD8+ T cells and their differentiation into cytotoxic effectors. However, secondary CTL expansion (that is, a secondary response upon re-encounter with antigen) is wholly dependent on the presence of T(H) cells during, but not after, priming. Our results demonstrate that T-cell help is 'programmed' into CD8+ T cells during priming, conferring on these cells a hallmark of immune response memory: the capacity for functional expansion on re-encounter with antigen.


Subject(s)
Immunologic Memory , Lymphocyte Activation , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigens/immunology , Cell Line , Cytotoxicity, Immunologic , Flow Cytometry , Lymphocytic choriomeningitis virus/immunology , Mice , T-Lymphocytes, Cytotoxic/cytology
16.
J Immunol ; 170(3): 1231-9, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12538681

ABSTRACT

Negative selection is designed to purge the immune system of high-avidity, self-reactive T cells and thereby protect the host from overt autoimmunity. In this in vivo viral infection model, we show that there is a previously unappreciated dichotomy involved in negative selection in which high-avidity CD8(+) T cells specific for a dominant epitope are eliminated, whereas T cells specific for a subdominant epitope on the same protein preferentially escape deletion. Although this resulted in significant skewing of immunodominance and a substantial depletion of the most promiscuous T cells, thymic and/or peripheral deletion of high-avidity CD8(+) T cells was not accompanied by any major change in the TCR V beta gene family usage or an absolute deletion of a single preferred complementarity-determining region 3 length polymorphism. This suggests that negative selection allows high-avidity CD8(+) T cells specific for subdominant or cryptic epitopes to persist while effectively deleting high-avidity T cells specific for dominant epitopes. By allowing the escape of subdominant T cells, this process still preserves a relatively broad peripheral TCR repertoire that can actively participate in antiviral and/or autoreactive immune responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Clonal Deletion/immunology , Epitopes, T-Lymphocyte/immunology , Immunodominant Epitopes/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/biosynthesis , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Clonal Deletion/genetics , Cytotoxicity, Immunologic/genetics , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Lymphocyte Activation/genetics , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Multigene Family/immunology , Nucleoproteins/biosynthesis , Nucleoproteins/genetics , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology , Viral Proteins/biosynthesis , Viral Proteins/genetics
17.
J Immunol ; 169(9): 4867-72, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12391197

ABSTRACT

In vivo administration of APC expressing Fas ligand (Fas-L(+) dendritic cells (DCs)) has shown promise in dampening allergic reactions and transplant rejection. Since the effect in these studies was mainly on CD4 lymphocytes, our goal was to evaluate the ability of such killer DCs to eliminate antiviral CD8 lymphocytes and in this way ameliorate viral immunopathology or, conversely, impede viral clearance. Intravenous administration of Fas-L(+) DCs resulted in a 50% reduction of lytic CD8 precursors following intracerebral infection with lymphocytic choriomeningitis virus (LCMV), and accordingly, immunopathology and survival of LCMV meningitis were improved, whereas viral clearance remained unaffected. In transfer studies the effect of the Fas-L(+) DCs was only quantifiable on experienced, not naive, CD8 lymphocytes. Importantly, loading of Fas-L(+) DCs with viral Ag before therapy was not necessary to achieve this effect, indicating that non-LCMV-infected Fas-L(+) DCs acquired viral Ag during acute LCMV infection in vivo. Our studies delineate important aspects for the clinical use of Fas-L(+) DCs in vivo. One should expect that they acquire viral Ags and suppress antiviral CD8 responses to some degree when given while an acute infection is ongoing. In terms of safety it is encouraging that resolution of the infection, at least in the case of LCMV, is not inhibited.


Subject(s)
Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/mortality , Membrane Glycoproteins/biosynthesis , fas Receptor/metabolism , Adoptive Transfer , Animals , Cell Line , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Dendritic Cells/virology , Fas Ligand Protein , Injections, Intravenous , Lymphocyte Activation , Lymphocyte Count , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/physiology , Mice , Mice, Inbred A , Mice, Inbred BALB C , Survival Analysis
18.
Immunity ; 16(3): 403-15, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11911825

ABSTRACT

Systemic treatment with antibody to CD40 ligand (aCD40L) can prevent autoimmunity and transplant rejection in several animal models and is currently under evaluation in clinical trials. While it is known that aCD40L administration inhibits expansion and effector functions of aggressive T cells, it is still unclear whether additional regulatory mechanisms are operative. Here we demonstrate that a single episode of CD40L blockade during development of the autoaggressive immune response completely prevented autoimmune disease in the RIP-LCMV mouse model for virally induced type 1 diabetes. Interestingly, protection could be transferred by a highly potent, bitypic cell population sharing phenotypic and functional properties of both natural killer (NK) and dendritic cells (DC). Furthermore, protection of prediabetic recipients was autoantigen specific and did not result in generalized immunosuppression. The origin, function, and therapeutic potential of these bitypic NK/DC regulatory cells is discussed.


Subject(s)
CD40 Ligand/immunology , Diabetes Mellitus, Type 1/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/prevention & control , Insulin/genetics , Killer Cells, Natural/immunology , Lymphocytic choriomeningitis virus , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic
19.
J Immunol ; 168(2): 933-41, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11777992

ABSTRACT

Treatment with nonmitogenic CD3 Ab reverses established autoimmune diabetes in nonobese diabetic mice by restoring self-tolerance, and is currently under clinical evaluation in patients presenting recent onset type I diabetes. Due to the immunosuppressive potential of this strategy, it was relevant to explore how this treatment would influence the outcome of concomitant viral infections. In this study, we used a transgenic model of virally induced autoimmune diabetes (rat insulin promoter-lymphocytic choriomeningitis virus) that allows for more precise tracking of the autoaggressive response and choice of the time point for initiation of autoimmunity. CD3 was most effective during a clearly defined prediabetic phase and prevented up to 100% of diabetes by drastically lowering activation of autoaggressive CD8 lymphocytes and their production of inflammatory cytokines. Interestingly, reversion of established disease could be achieved as well, when nonmitogenic CD3 was administered late during pathogenesis to overtly diabetic recipients. Most importantly, competence to clear viral infections was maintained. Thus, administration of nonmitogenic CD3 prevents diabetes by sufficient systemic reduction of (auto)aggressive lymphocytes, but without compromising antiviral immune competence.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD3 Complex/immunology , Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 1/virology , Insulin/genetics , Lymphocytic choriomeningitis virus/genetics , Promoter Regions, Genetic/immunology , Adjuvants, Immunologic/pharmacology , Adoptive Transfer , Animals , Antibodies, Monoclonal/metabolism , Arenaviridae Infections/immunology , Arenaviridae Infections/prevention & control , Arenaviridae Infections/virology , Binding Sites, Antibody , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Division/immunology , Cell Movement/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fab Fragments/pharmacology , Insulin/immunology , Interleukin-4/biosynthesis , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphotoxin-alpha/antagonists & inhibitors , Lymphotoxin-beta , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogens/pharmacology , Rats , Receptors, Fc/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Spleen/transplantation , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Viral Proteins/genetics
20.
Eur J Immunol ; 32(1): 113-21, 2002 01.
Article in English | MEDLINE | ID: mdl-11754351

ABSTRACT

Administration of autoantigens through DNA immunizations or via the oral route can prevent progression of islet destruction and lower the incidence of type 1 diabetes in animal models. This beneficial effect is mediated by autoreactive regulatory CD4 lymphocytes, and it is known that their induction depends on the precise dose and route of antigen administration. However, it is not clear which endogenous factors determine when such immunizations lead to activation of regulatory versus aggressive autoreactive lymphocytes and how a deleterious outcome can be avoided. Here we describe novel observations made in an animal model for virally induced type 1 diabetes, showing that the endogenous expression levels of the islet antigens and glutamic acid decarboxylase determine whether immunization with these antigens is beneficial or detrimental. Lower expression levels in beta-cells support immune regulation resulting in induction of autoreactive, regulatory cells characterized by increased IL-4 production (Th2-like), whereas higher levels favor Th1-like autoaggressive responses characterized by augmented IFN-gamma generation. Co-immunization with an IL-4-expressing plasmid reduces the risk of augmenting autoaggression and in this way increases the safety margin of this immune-based therapy. Our findings will be of importance for designing safe antigen-specific interventions for human type 1 diabetes.


Subject(s)
Autoantigens/immunology , Diabetes Mellitus, Type 1/prevention & control , Glutamate Decarboxylase/immunology , Interleukin-4/immunology , Isoenzymes/immunology , Vaccines, DNA/immunology , Animals , Antigens, Viral/genetics , Autoantibodies/biosynthesis , Autoantigens/genetics , Cell Line , Chlorocebus aethiops , Consumer Product Safety , Cricetinae , Diabetes Mellitus, Type 1/physiopathology , Female , Gene Expression , Glutamate Decarboxylase/genetics , Glycoproteins/genetics , Humans , Insulin/metabolism , Interleukin-4/genetics , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Isoenzymes/genetics , Lymphocytic choriomeningitis virus , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Plasmids , Th1 Cells/immunology , Th2 Cells/immunology , Treatment Outcome , Vaccination , Vaccines, DNA/genetics , Vero Cells , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...