Project description:We previously generated the Orex-HA mouse model in which orexin-producing neurons residing in the hypothalamus selectively expressed a model self-antigen, namely hemagglutinin (HA) from the H1N1 influenza virus. In this model the adoptive transfer of in vitro activated HA-specific CD8 T cells leads to the specific loss of orexinergic neurons located in the hypothalamus. To assess the phenotype and investigate the pathogenic mechanisms and the potential tissue-resident properties of autoreactive (HA-specific) CD8 T cells infiltrating the hypothalamus of Orex-HA mice , we performed a single-cell transcriptomic analysis (scRNA-seq) of HA-specific CD8+ T cells isolated from the hypothalamus and the spleen at day 30 after T cell transfer in Orex-HA mice.
Project description:The precise timing and pathway of memory CD8+ T cells differentiation from naïve T cells have remained undetermined. We found the smaller cell-size and slower cell cycling cells were segregated from the proliferative larger cell-size activated T cell pool at the peak of infection. Gene signature of the smaller cell-size slower cycling cells and the large cell-size proliferative cells was compared to the signature of naïve, effector, central and effector memory CD8+ T cells. Total RNA samples were prepared from sorted populations of larger or smaller cell-sized cells from spleens of influenza virus PR8-OVA-infected mice on day 7 p.i. or from in vitro 7 days culture after stimulation with plate-bound anti-CD3ε (1.0 μg ml−1) and anti-CD28 mAb (0.5 μg ml−1). Effector T-cell control samples were prepared from SIINFEKL (100 ng ml−1) stimulated OT-I cells after 4 days of in vitro culture with rIL-2 (10 ng/ml) and sorted as CD8+CD44hiCD62Llo. Control bona fide effector memory and central memory T cells were sorted from the spleens of PR8-OVA-infected mice on day 42 p.i. Naive cells were sorted as CD8+CD44loCD62Lhi cells from uninfected C57BL/6 mice.
Project description:Influenza virus infection-induced gene expression changes of regional B cells are mediated at least in part through type I Interferon:; Our objective is to determine whether the influenza virus-infection induced gene expression changes in regional lymph node B cells are facilitated at least in part through type I interferon. Our specific aim is to compare the gene expression profile of highly FACS-purified B cells in the regional lymph nodes of wildtype and IFNR-/- mice prior to and 48h following infection with influenza virus infection and to contrast this expression profile with that of FACS-purified wildtype B cells activated in vitro with IFN-beta anti-CD86 for 12h. Experiment Overall Design: We analyzed gene expression from mouse lymph node B cells purified by flow cytometric sorting using single channel oligonucleotide microarrays. There were 4 groups: 1) wild type uninfected mice (control group), 2) wild type mice infected with influenza (flu) for 2 days, 3) IFNR-/- mice infected with flu for 2 days, 4) cells stimulated with IFN-b in vitro for 17 h. Each group contained 4 biological replicates obtained from independent experiments. There were 16 total samples and each was measured on a separate array.
Project description:Activation of peripheral T lymphocytes in the absence of pathogen induced inflammation or costimulatory signals results in tolerance. Two different tolerance mechanisms have been described, deletion and anergy. We recently demonstrated that an important variable which is responsible for the decision between anergy and deletion for CD8 T cells is the strength of signaling through the T cell receptor(Redmond and Sherman, Immunity 22:275,2005). However, it is not know what the downstream signals are that result in death(deletion) vs. survival(anergy)of the activated cells. Here we analysze additional conditions: 1) Wild Type mice were injected injected only once with a strain of vaccinia virus that express hemagglutinin from influenza; 2) BIM KO Clone 4 transgenic T cells were transferred into B10D2 mice then injected daily for 3 days with 1ug of KdHA peptide and sorted by flow cytometry according to their expression of CD8 and thy1.1; 3) Mice injected with 1 ug (deleted condition) of influenza hemaglutinin antigen (HA), 24 hours post injection CD8 T CL4 cells were sorted by flow cytometry (FacsARIA) and 4) WildType mice injected with 100 ug (anergic condition) of influenza hemaglutinin antigen (HA), 24 hours post injection CD8 T CL4 cells were sorted by flow cytometry (FacsARIA). Dr. Sherman's lab aims to assess the role of protein glycosylation in the decision between deletion vs. anergy in immune tolerance, they have prepared purified TCR transgenic CD8 T cells stimulated in vivo using a strong antigenic signal (anergy conditions) or a weak antigenic signal (deletion conditions) with cognate antigen. Dr. Sherman's lab wishes to assess transcriptional differences in genes involved in protein glycosylation in these 2 cell populations. Then control population will be naïve transgenic CD8 cells. RNA preparations of mouse CD8 T cells from the B10D2 strain with three different conditions (Naïve, Deleted, and Anergic) were sent to the Microarray Core (E). The RNA was amplified, labeled, and hybridized to the GLYCOv4 microarrays.
Project description:MIcroRNA expression profiling of primary murine splenic dendritic cells (Flt3L expanded) comparing untreated cells to cells infected with Influenza A or stimulated with polyI:C in vitro. Three-condition experiment, Influenza A/PR/8/34 virus MOI=10 vs polyI:C 30 ug/mL vs. mock treated; RNA collected after 8 hours. 3 Independent biological replicates from cells prepared on different days. One replicate per array.
Project description:Role for naturally occurring CD4+CD25+Foxp3+ regulatory T cells (nTregs) in counterbalancing this process. Using a transgenic murine model for autoimmune-mediated lung disease, we demonstrated that, despite pulmonary inflammation, lung-specific CD8+ T cells can reside quiescently in close proximity to self-antigen. Whereas self-reactive CD8+ T cells in the inflamed lung and lung-draining lymph nodes down-regulated the expression of effector molecules, those located in the spleen appeared to be partly antigen-experienced and displayed a memory-like phenotype. Since ex vivo-reisolated self-reactive CD8+ T cells were very well capable to respond to the antigen in vitro, we investigated a possible contribution of nTregs to the immune control over autoaggressive CD8+ T cells in the lung. We isolated antigen-specifc CD8+ T-cells from lungs and bronchial lymphnodes derived from chronic diseased mice (SPC-HAxCL4), healthy control mice (CL4) and acute influenza infected control mice (CL4+IAV) and perormed mRNA expression profiling of isolated CD8+ T cells. Each group represents a pool of at least n=4 animals. CD8+ T cell type comparison; lung disease state analysis
Project description:Analysis of the gene expression profiles of naïve B cells, resting memory B cells (MBCs), activated B cells (ABCs) and antibody-secreting cells (ASCs) isolated from peripheral blood one week following influenza vaccination. Upon antigen exposure B cells eventually bifurcate into two distinct lineages, plasmablasts and memory B cells. We previously reported that plasmablasts or antibody-secreting cells (ASCs) could be transiently detected in blood shortly after infection or vaccination of humans. Here, we define the phenotype and the transcriptional program of a novel human antigen-specific B cell subset, referred to as activated B cells (ABCs). ABCs do not spontaneously secrete antibodies and possess a unique transcriptional profile that distinguishes them from ASCs and resting memory B cells. Clonal lineages present among day 7 ABCs persisted in blood for up to three months post-influenza immunization indicating that ABCs may be destined to join the long-term memory B cell pool. ABCs and ASCs can be clearly distinguished in blood following influenza and Ebola virus infections. Interrogating ABCs will expand our understanding of the differentiation, maturation and longevity of human B cell responses. Total RNA was isolated from 10,000 cells of each population.
Project description:We speculated that distinct levels of Id2 was deterministic in the transcriptional program of antigen-specific CD8+ T cells. To test this hypothesis, we subjected DbNP366-specific effector CD8+ T cells purified according to their differential expression of Id2-GFP (Id2-GFPint and Id2-GFPhigh) to microarray analysis and compared their gene expression profiles to the differentially expressed genes identified by comparing Id2-deficient and wild-type DbNP366-specific CD8+ T cells. This analysis revealed that the transcriptional program of CD8+ T cell differentiation is exquisitely sensitive to the concentration of Id2. PR8-primed Id2gfp/gfp mice were challenged after one month with influenza intranasally HKx31 virus and analysed on day 9 for the expression of Id2-GFP in DbNP366+CD44+ CD8+ T cells. DbNP366-specific CD8+ T cells were then separated into virus-specific CD8+ T cells that expressed intermediate or high levels of GFP. Purified Id2-GFPintermediate (int) and Id2-GFPhigh DbNP366-specific CD8+ T cells were analysed by mRNA whole genome microarray. Three replicates of each group (Id2-GFPint or Id2-GFPhigh) were analysed.
Project description:In comparison to murine dendritic cells (DCs), less is known about the function of human DCs in tissues. Here, we analyzed, using lung tissues from humans and humanized mice, the role of human CD1c+ and CD141+ DCs in determining the type of CD8+ T cell immunity to live-attenuated influenza virus (LAIV) vaccine. We found that both lung DC subsets acquired influenza antigens in vivo and expanded specific cytotoxic CD8+ T cells in vitro. However, lung-tissue-resident CD1c+ DCs but not CD141+ DCs were able to drive CD103 expression on CD8+ T cells and promote CD8+ T cell accumulation in lung epithelia in vitro and in vivo. CD1c+ DCs induction of CD103 expression was dependent on membrane-bound TGF-?1. Thus, CD1c+ and CD141+ DCs generate CD8+ T cells with different properties, and CD1c+ DCs specialize in the regulation of mucosal CD8+ T cells. Total RNA were isolated from purified human CD1c+ (BDCA1+) and CD141+ (BDCA3+) mDCs sorted from different tissues, including human blood, spleen and lungs of humanized mice, and human lungs. Eighteen samples in total were analyzed from different donors and tissues.
Project description:Vaccine development involves time-consuming and expensive evaluation of candidate vaccines in animal models. As mediators of both innate and adaptive immune responses dendritic cells (DCs) are considered to be highly important for vaccine performance. Here we evaluated in how far the response of DCs to a vaccine in vitro is in line with the immune response the vaccine evokes in vivo. To this end, we investigated the response of murine bone marrow-derived DCs to whole inactivated virus (WIV) and subunit (SU) influenza vaccine preparations. These vaccine preparations were chosen because they differ in the immune response they evoke in mice with WIV being superior to SU vaccine through induction of higher virus-neutralizing antibody titers and a more favorable Th1-skewed response phenotype. To evaluate if in vivo immunogenicity is reflected by DC reactions in vitro we studied the gene expression signature of murine bone-marrow-derived conventional DCs (cDCs) upon stimulation with WIV or SU influenza vaccine or, for reasons of comparison, with live influenza virus. Dendritic cells stimulated with PBS served as a control. Gene expression analysis was performed on samples 4, 12 and 24 hours after the start of stimulation.