Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cytometry B Clin Cytom ; 98(2): 146-160, 2020 03.
Article in English | MEDLINE | ID: mdl-31758746

ABSTRACT

High-dimensional mass cytometry data potentially enable a comprehensive characterization of immune cells. In order to positively affect clinical trials and translational clinical research, this advanced technology needs to demonstrate a high reproducibility of results across multiple sites for both peripheral blood mononuclear cells (PBMC) and whole blood preparations. A dry 30-marker broad immunophenotyping panel and customized automated analysis software were recently engineered and are commercially available as the Fluidigm® Maxpar® Direct™ Immune Profiling Assay™. In this study, seven sites received whole blood and six sites received PBMC samples from single donors over a 2-week interval. Each site labeled replicate samples and acquired data on Helios™ instruments using an assay-specific acquisition template. All acquired sample files were then automatically analyzed by Maxpar Pathsetter™ software. A cleanup step eliminated debris, dead cells, aggregates, and normalization beads. The second step automatically enumerated 37 immune cell populations and performed label intensity assessments on all 30 markers. The inter-site reproducibility of the 37 quantified cell populations had consistent population frequencies, with an average %CV of 14.4% for whole blood and 17.7% for PBMC. The dry reagent coupled with automated data analysis is not only convenient but also provides a high degree of reproducibility within and among multiple test sites resulting in a comprehensive yet practical solution for deep immune phenotyping.


Subject(s)
Blood Cells/cytology , Flow Cytometry , Immunophenotyping , Automation, Laboratory/instrumentation , Automation, Laboratory/methods , Automation, Laboratory/standards , Canada , Data Analysis , Flow Cytometry/instrumentation , Flow Cytometry/methods , Flow Cytometry/standards , Humans , Immunophenotyping/instrumentation , Immunophenotyping/methods , Immunophenotyping/standards , Laboratory Proficiency Testing , Leukocytes, Mononuclear/cytology , Pattern Recognition, Automated/methods , Pattern Recognition, Automated/standards , Reference Standards , Reproducibility of Results , United States
2.
J Vis Exp ; (117)2016 11 16.
Article in English | MEDLINE | ID: mdl-27911410

ABSTRACT

L. monocytogenes is a gram-positive bacterium that is a cause of food borne disease in humans. Experimental infection of mice with this pathogen has been highly informative on the role of innate and adaptive immune cells and specific cytokines in host immunity against intracellular pathogens. Production of IFN-γ by innate cells during sublethal infection with L. monocytogenes is important for activating macrophages and early control of the pathogen1-3. In addition, IFN-γ production by adaptive memory lymphocytes is important for priming the activation of innate cells upon reinfection4. The L. monocytogenes infection model thus serves as a great tool for investigating whether new therapies that are designed to increase IFN-γ production have an impact on IFN-γ responses in vivo and have productive biological effects such as increasing bacterial clearance or improving mouse survival from infection. Described here is a basic protocol for how to conduct intraperitoneal infections of C57BL/6J mice with the EGD strain of L. monocytogenes and to measure IFN-γ production by NK cells, NKT cells, and adaptive lymphocytes by flow cytometry. In addition, procedures are described to: (1) grow and prepare the bacteria for inoculation, (2) measure bacterial load in the spleen and liver, and (3) measure animal survival to endpoints. Representative data are also provided to illustrate how this infection model can be used to test the effect of specific agents on IFN-γ responses to L. monocytogenes and survival of mice from this infection.


Subject(s)
Host-Pathogen Interactions , Immunity, Innate , Interferon-gamma , Listeria monocytogenes , Listeriosis , Animals , Mice , Mice, Inbred C57BL
3.
J Immunol ; 197(11): 4464-4472, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27799307

ABSTRACT

NKT cells are unconventional T cells that respond to self and microbe-derived lipid and glycolipid Ags presented by the CD1d molecule. Invariant NKT (iNKT) cells influence immune responses in numerous diseases. Although only a few studies have examined their role during intestinal inflammation, it appears that iNKT cells protect from Th1-mediated inflammation but exacerbate Th2-mediated inflammation. Studies using iNKT cell-deficient mice and chemically induced dextran sodium sulfate (DSS) colitis have led to inconsistent results. In this study, we show that CD1d-deficient mice, which lack all NKT cells, harbor an altered intestinal microbiota that is associated with exacerbated intestinal inflammation at steady-state and following DSS treatment. This altered microbiota, characterized by increased abundance of the bacterial phyla Proteobacteria, Deferribacteres, and TM7, among which the mucin-eating Mucispirillum, as well as members of the genus Prevotella and segmented filamentous bacteria, was transmissible upon fecal transplant, along with the procolitogenic phenotype. Our results also demonstrate that this proinflammatory microbiota influences iNKT cell function upon activation during DSS colitis. Collectively, alterations of the microbiota have a major influence on colitis outcome and therefore have to be accounted for in such experimental settings and in studies focusing on iNKT cells.


Subject(s)
Colitis/immunology , Colitis/microbiology , Gastrointestinal Microbiome/immunology , Lymphocyte Activation , Natural Killer T-Cells/immunology , Animals , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate/toxicity , Fecal Microbiota Transplantation , Inflammation/chemically induced , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Mice , Mice, Knockout , Natural Killer T-Cells/pathology , Prevotella/immunology , Th2 Cells/immunology , Th2 Cells/pathology
4.
J Immunol ; 195(11): 5189-202, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26491197

ABSTRACT

Females exhibit more robust Th1 responses than males. Our previous work suggested that this sex disparity is a consequence of higher activity of the androgen-induced gene peroxisome proliferator-activated receptor α (PPARα) in male CD4(+) T cells. The objective of this study was to elucidate the cellular and molecular mechanism of how PPARα inhibits Th1 responses in male mice. In this study, we found that PPARα functions within CD4(+) and CD8(+) T lymphocytes and NKT cells to negatively regulate IFN-γ responses in male mice and identified Ifng as the gene target of PPARα repression. Treatment of male CD4(+) T cells with the PPARα agonist fenofibrate induced the recruitment of PPARα and the nuclear receptor-interacting protein, nuclear receptor corepressor 1, to specific cis-regulatory elements in the Ifng locus. This recruitment associated with reduced histone acetylation at these sites. Knockdown of nuclear receptor corepressor 1 in primary male T cells abolished the effect of fenofibrate in reducing IFN-γ production. In contrast, treatment of male T cells with IS001, a novel antagonist of PPARα, increased Ifng gene expression and histone acetylation across the Ifng locus. Finally, we investigated the effects of IS001 on IFN-γ responses in mice during infection with the Th1-associated pathogen Listeria monocytogenes and observed that IS001 enhanced IFN-γ production by NKT, CD4(+), and CD8(+) T cells and improved the survival of male, but not female, mice. Our findings provide a novel mechanism of why IFN-γ responses are more robust in females and introduce a small-molecule IS001 that can be used to enhance Th1 immunity in males.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Natural Killer T-Cells/immunology , PPAR alpha/physiology , Th1 Cells/immunology , Acetylation , Acrylamides/pharmacology , Animals , Fenofibrate/pharmacology , Histones/metabolism , Interferon-gamma/biosynthesis , Listeria monocytogenes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , PPAR alpha/antagonists & inhibitors , PPAR alpha/genetics , Pyridinium Compounds/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Sex Factors
5.
J Immunol ; 191(11): 5646-54, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24163408

ABSTRACT

Invariant NKT (iNKT) cells act at the crossroad between innate and adaptive immunity and are important players in the defense against microbial pathogens. iNKT cells can detect pathogens that trigger innate receptors (e.g., TLRs, Rig-I, Dectin-1) within APCs, with the consequential induction of CD1d-mediated Ag presentation and release of proinflammatory cytokines. We show that the cytosolic peptidoglycan-sensing receptors Nod1 and Nod2 are necessary for optimal IFN-γ production by iNKT cells, as well as NK cells. In the absence of Nod1 and Nod2, iNKT cells had a blunted IFN-γ response following infection by Salmonella enterica serovar Typhimurium and Listeria monocytogenes. For Gram-negative bacteria, we reveal a synergy between Nod1/2 and TLR4 in dendritic cells that potentiates IL-12 production and, ultimately, activates iNKT cells. These findings suggest that multiple innate pathways can cooperate to regulate iNKT cell activation during bacterial infection.


Subject(s)
Listeria monocytogenes/immunology , Listeriosis/immunology , Natural Killer T-Cells/immunology , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Salmonella typhi/immunology , Typhoid Fever/immunology , Animals , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/microbiology , Immunity, Innate/genetics , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , Toll-Like Receptor 4/metabolism
6.
Curr Opin Immunol ; 24(4): 398-404, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22677577

ABSTRACT

The Nod-like receptor (NLR) family of intracellular pattern recognition molecules plays critical roles in the control of inflammation through the modulation of different signalling pathways, including those dependent on NF-κB and caspase-1-mediated cleavage of interleukin (IL)-1ß and IL-18. A number of NLRs or NLR-associated proteins have been genetically associated with susceptibility to inflammatory bowel disease (IBD), either Crohn's disease or ulcerative colitis. Accordingly, recent studies have examined the role of NLR proteins in chemical-induced or bacteria-induced murine models of colitis. In this review, we will discuss the genetic associations of NLRs with IBD and the research using NLR-deficient mice in different colitis models.


Subject(s)
Colitis, Ulcerative/immunology , Crohn Disease/immunology , Nod1 Signaling Adaptor Protein/physiology , Nod2 Signaling Adaptor Protein/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Humans , Mice
7.
Immunology ; 136(3): 273-82, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22671023

ABSTRACT

Invariant natural killer T (iNKT) cells are evolutionarily conserved lipid-reactive T cells that bridge innate and adaptive immune responses. Despite a relatively restricted T-cell receptor (TCR) diversity, these cells respond to a variety of structurally distinct foreign (i.e. microbial or synthetic) as well as host-derived (self-) lipid antigens presented by the CD1d molecule. These multi-tasking lymphocytes are among the first responders in immunity, and produce an impressive array of cytokines and chemokines that can tailor the ensuing immune response. Accordingly, iNKT cells play important functions in autoimmune diseases, cancer, infection and inflammation. These properties make iNKT cells appealing targets in immune-based therapies. Yet, much has to be learned on the mechanisms that allow iNKT cells to produce polarized responses. Responses of iNKT cells are influenced by the direct signals perceived by the cells through their TCRs, as well as by indirect co-stimulatory (and potentially co-inhibitory) cues that they receive from antigen-presenting cells or the local milieu. A decade ago, biochemists and immunologists have started to describe synthetic lipid agonists with cytokine skewing potential, paving a new research avenue in the iNKT cell field. Yet how iNKT cells translate various antigenic signals into distinct functional responses has remained obscure. Recent findings have revealed a unique and innate mode of lipid recognition by iNKT cells, and suggest that both the lipid antigen presented and the diversity of the TCR modulate the strength of CD1d-iNKT TCR interactions. In this review, we focus on novel discoveries on lipid recognition by iNKT cells, and how these findings may help us to design effective strategies to steer iNKT cell responses for immune intervention.


Subject(s)
Lipids/immunology , Natural Killer T-Cells/immunology , Animals , Antigens/metabolism , Antigens, CD1d/metabolism , Chemokines/biosynthesis , Cytokines/biosynthesis , Humans , Immunity, Innate , Lymphocyte Activation , Models, Immunological , Natural Killer T-Cells/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology
8.
Proc Natl Acad Sci U S A ; 108(36): 14896-901, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21856952

ABSTRACT

Although a number of studies have examined the development of T-helper cell type 2 (Th2) immunity in different settings, the mechanisms underlying the initiation of this arm of adaptive immunity are not well understood. We exploited the fact that immunization with antigen plus either nucleotide-binding oligomerization domain-containing proteins 1 (Nod1) or 2 (Nod2) agonists drives Th2 induction to understand how these pattern-recognition receptors mediate the development of systemic Th2 immune responses. Here, we show in bone-marrow chimeric mice that Nod1 and Nod2 expression within the stromal compartment is necessary for priming of effector CD4(+) Th2 responses and specific IgG1 antibodies. In contrast, sensing of these ligands by dendritic cells was not sufficient to induce Th2 immunity, although these cells contribute to the response. Moreover, we determined that CD11c(+) cells were the critical antigen-presenting cells, whereas basophils and B cells did not affect the capacity of Nod ligands to induce CD4(+) Th2 effector function. Finally, we found that full Th2 induction upon Nod1 and Nod2 activation was dependent on both thymic stromal lymphopoietin production by the stromal cells and the up-regulation of the costimulatory molecule, OX40 ligand, on dendritic cells. This study provides in vivo evidence of how systemic Th2 immunity is induced in the context of Nod stimulation. Such understanding will influence the rational design of therapeutics that could reprogram the immune system during an active Th1-mediated disease, such as Crohn's disease.


Subject(s)
Cytokines/immunology , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Th2 Cells/immunology , Animals , B-Lymphocytes/immunology , Basophils/immunology , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/therapy , Cytokines/genetics , Dendritic Cells/immunology , Immunity, Cellular/physiology , Immunization , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , OX40 Ligand , Protein Structure, Tertiary , Th1 Cells/immunology , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/immunology , Thymic Stromal Lymphopoietin
9.
J Neural Transm (Vienna) ; 118(5): 795-808, 2011 May.
Article in English | MEDLINE | ID: mdl-21552986

ABSTRACT

Sequence variants at or near the leucine-rich repeat kinase 2 (LRRK2) locus have been associated with susceptibility to three human conditions: Parkinson's disease (PD), Crohn's disease and leprosy. As all three disorders represent complex diseases with evidence of inflammation, we hypothesized a role for LRRK2 in immune cell functions. Here, we report that full-length Lrrk2 is a relatively common constituent of human peripheral blood mononuclear cells (PBMC) including affinity isolated, CD14(+) monocytes, CD19(+) B cells, and CD4(+) as well as CD8(+) T cells. Up to 26% of PBMC from healthy donors and up to 43% of CD14(+) monocytes were stained by anti-Lrrk2 antibodies using cell sorting. PBMC lysates contained full-length (>260 kDa) and higher molecular weight Lrrk2 species. The expression of LRRK2 in circulating leukocytes was confirmed by microscopy of human blood smears and in sections from normal midbrain and distal ileum. Lrrk2 reactivity was also detected in mesenteric lymph nodes and spleen (including in dendritic cells), but was absent in splenic mononuclear cells from lrrk2-null mice, as expected. In cultured bone marrow-derived macrophages from mice we made three observations: (i) a predominance of higher molecular weight lrrk2; (ii) the reduction of autophagy marker LC3-II in (R1441C)lrrk2-mutant cells (<31%); and (iii) a significant up-regulation of lrrk2 mRNA (>fourfold) and protein after exposure to several microbial structures including bacterial lipopolysaccharide and lentiviral particles. We conclude that Lrrk2 is a constituent of many cell types in the immune system. Following the recognition of microbial structures, stimulated macrophages respond with altered lrrk2 gene expression. In the same cells, lrrk2 appears to co-regulate autophagy. A pattern recognition receptor-type function for LRRK2 could explain its locus' association with Crohn's disease and leprosy risk. We speculate that the role of Lrrk2 in immune cells may also be relevant to the susceptibility of developing PD or its progression.


Subject(s)
Genetic Predisposition to Disease/genetics , Leukocytes/metabolism , Macrophages/metabolism , Parkinson Disease , Protein Serine-Threonine Kinases/genetics , Up-Regulation/physiology , Animals , Antibody-Dependent Cell Cytotoxicity , Autophagy/genetics , B-Lymphocytes/metabolism , Cells, Cultured , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Mice, Transgenic , Mutation/genetics , Parkinson Disease/genetics , Parkinson Disease/immunology , Parkinson Disease/pathology , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , T-Lymphocytes/metabolism
10.
Infect Immun ; 78(12): 5107-15, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20921147

ABSTRACT

The pattern recognition molecules Nod1 and Nod2 play important roles in intestinal homeostasis; however, how these proteins impact on the development of inflammation during bacterial colitis has not been examined. In the streptomycin-treated mouse model of Salmonella colitis, we found that mice deficient for both Nod1 and Nod2 had attenuated inflammatory pathology, reduced levels of inflammatory cytokines, and increased colonization of the mucosal tissue. Nod1 and Nod2 from both hematopoietic and nonhematopoietic sources contributed to the pathology, and all phenotypes were recapitulated in mice deficient for the signaling adaptor protein Rip2. However, the influence of Rip2 was strictly dependent on infection conditions that favored expression of the Salmonella pathogenicity island 2 (SPI-2) type III secretion system (TTSS), as Rip2 was dispensable for inflammation when mice were infected with bacteria grown under conditions that promoted expression of the SPI-1 TTSS. Thus, Nod1 and Nod2 can modulate inflammation and mediate efficient clearance of bacteria from the mucosal tissue during Salmonella colitis, but their role is dependent on the expression of the SPI-2 TTSS.


Subject(s)
Colitis/microbiology , Nod1 Signaling Adaptor Protein/physiology , Nod2 Signaling Adaptor Protein/physiology , Salmonella Infections, Animal/immunology , Animals , Bacterial Secretion Systems/immunology , Bacterial Secretion Systems/physiology , Chemokines/physiology , Colitis/immunology , Colitis/physiopathology , Enzyme-Linked Immunosorbent Assay , Interleukin-1beta/physiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Salmonella Infections, Animal/physiopathology
11.
Infect Immun ; 77(10): 4480-6, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19620349

ABSTRACT

Recent advances in immunology have highlighted the critical function of pattern-recognition molecules (PRMs) in generating the innate immune response to effectively target pathogens. Nod1 and Nod2 are intracellular PRMs that detect peptidoglycan motifs from the cell walls of bacteria once they gain access to the cytosol. Salmonella enterica serovar Typhimurium is an enteric intracellular pathogen that causes a severe disease in the mouse model. This pathogen resides within vacuoles inside the cell, but the question of whether cytosolic PRMs such as Nod1 and Nod2 could have an impact on the course of S. Typhimurium infection in vivo has not been addressed. Here, we show that deficiency in the PRM Nod1, but not Nod2, resulted in increased susceptibility toward a mutant strain of S. Typhimurium that targets directly lamina propria dendritic cells (DCs) for its entry into the host. Using this bacterium and bone marrow chimeras, we uncovered a surprising role for Nod1 in myeloid cells controlling bacterial infection at the level of the intestinal lamina propria. Indeed, DCs deficient for Nod1 exhibited impaired clearance of the bacteria, both in vitro and in vivo, leading to increased organ colonization and decreased host survival after oral infection. Taken together, these findings demonstrate a key role for Nod1 in the host response to an enteric bacterial pathogen through the modulation of intestinal lamina propria DCs.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/microbiology , Nod1 Signaling Adaptor Protein/immunology , Salmonella Infections/immunology , Salmonella typhimurium/growth & development , Salmonella typhimurium/immunology , Animals , Colony Count, Microbial , Gene Deletion , Liver/microbiology , Lymph Nodes/microbiology , Mice , Mice, Inbred C57BL , Nod1 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/immunology , Salmonella Infections/microbiology , Spleen/microbiology , Survival Analysis
12.
J Immunol ; 181(11): 7925-35, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017983

ABSTRACT

While a number of microbial-associated molecular patterns have been known for decades to act as adjuvants, the mechanisms and the signaling pathways underlying their action have remained elusive. Here, we examined the unfolding of the adaptive immune response induced by Nod2 in vivo upon activation by its specific ligand, muramyl dipeptide, a component of peptidoglycan. Our findings demonstrate that this bacterial sensor triggers a potent Ag-specific immune response with a Th2-type polarization profile, characterized by the induction of IL-4 and IL-5 by T cells and IgG1 Ab responses. Nod2 was also found to be critical for the induction of both Th1- and Th2-type responses following costimulation with TLR agonists. Importantly, the synergistic responses to Nod2 and TLR agonists seen in vivo were recapitulated by dendritic cells in vitro, suggesting that these cells likely play a central role in the integration of Nod2- and TLR-dependent signals for driving the adaptive immune response. Taken together, our results identify Nod2 as a critical mediator of microbial-induced potentiation and polarization of Ag-dependent immunity. Moreover, these findings affect our understanding of Crohn's diseases pathogenesis, where lack of Nod2-dependent Th2 signaling in a subset of these patients might explain heightened Th1-mediated inflammation at the level of the intestinal mucosa.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/immunology , Antigens, Bacterial/immunology , Nod2 Signaling Adaptor Protein/immunology , Peptidoglycan/immunology , Signal Transduction/immunology , Th2 Cells/immunology , Toll-Like Receptors/immunology , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Animals , Antigens, Bacterial/pharmacology , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/pathology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-5/genetics , Interleukin-5/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Nod2 Signaling Adaptor Protein/genetics , Peptidoglycan/pharmacology , Signal Transduction/genetics , Th1 Cells/immunology , Th1 Cells/pathology , Th2 Cells/pathology , Toll-Like Receptors/genetics
13.
Diabetes ; 57(4): 918-28, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18198358

ABSTRACT

OBJECTIVE: Type 1 diabetes reflects autoimmune destruction of beta-cells and peri-islet Schwann cells (pSCs), but the mechanisms of pSC death and the T-cell epitopes involved remain unclear. RESEARCH DESIGN AND METHODS: Primary pSC cultures were generated and used as targets in cytotoxic T-lymphocyte (CTL) assays in NOD mice. Cognate interaction between pSC and CD8(+) T-cells was assessed by transgenic restoration of beta2-microglobulin (beta2m) to pSC in NOD.beta2m(-/-) congenics. I-A(g7) and K(d) epitopes in the pSC antigen glial fibrillary acidic protein (GFAP) were identified by peptide mapping or algorithms, respectively, and the latter tested by immunotherapy. RESULTS: pSC cultures did not express major histocompatibility complex (MHC) class II and were lysed by ex vivo CTLs from diabetic NOD mice. In vivo, restoration of MHC class I in GFAP-beta2m transgenics significantly accelerated adoptively transferred diabetes. Target epitopes in the pSC autoantigen GFAP were mapped to residues 79-87 and 253-261 for K(d) and 96-110, 116-130, and 216-230 for I-A(g7). These peptides were recognized spontaneously in NOD spleens as early as 2.5 weeks of age, with proliferative responses peaking around weaning and detectable lifelong. Several were also recognized by T-cells from new-onset type 1 diabetic patients. NOD mouse immunotherapy at 8 weeks with the CD8(+) T-cell epitope, GFAP 79-87 but not 253-261, significantly inhibited type 1 diabetes and was associated with reduced gamma-interferon production to whole protein GFAP. CONCLUSIONS: Collectively, these findings elucidate a role for pSC-specific CD8(+) T-cells in islet inflammation and type 1 diabetes pathogenesis, further supporting neuronal involvement in beta-cell demise.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Animals , Diabetes Mellitus, Type 1/pathology , Female , Glial Fibrillary Acidic Protein/genetics , Insulin/genetics , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Ovulation , Schwann Cells/pathology , T-Lymphocytes, Cytotoxic/immunology , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...