13 research outputs found

    The thymocyte-specific RNA-binding protein Arpp21 provides TCR repertoire diversity by binding to the 3'-UTR and promoting Rag1 mRNA expression

    Get PDF
    The regulation of thymocyte development by RNA-binding proteins (RBPs) is largely unexplored. We identify 642 RBPs in the thymus and focus on Arpp21, which shows selective and dynamic expression in early thymocytes. Arpp21 is downregulated in response to T cell receptor (TCR) and Ca2+ signals. Downregulation requires Stim1/Stim2 and CaMK4 expression and involves Arpp21 protein phosphorylation, polyubiquitination and proteasomal degradation. Arpp21 directly binds RNA through its R3H domain, with a preference for uridine-rich motifs, promoting the expression of target mRNAs. Analysis of the Arpp21-bound transcriptome reveals strong interactions with the Rag1 3 '-UTR. Arpp21-deficient thymocytes show reduced Rag1 expression, delayed TCR rearrangement and a less diverse TCR repertoire. This phenotype is recapitulated in Rag1 3 '-UTR mutant mice harboring a deletion of the Arpp21 response region. These findings show how thymocyte-specific Arpp21 promotes Rag1 expression to enable TCR repertoire diversity until signals from the TCR terminate Arpp21 and Rag1 activities

    Tumor Invasion of Salmonella enterica Serovar Typhimurium Is Accompanied by Strong Hemorrhage Promoted by TNF-α

    Get PDF
    BACKGROUND:Several facultative anaerobic bacteria with potential therapeutic abilities are known to preferentially colonize solid tumors after systemic administration. How they efficiently find and invade the tumors is still unclear. However, this is an important issue to be clarified when bacteria should be tailored for application in cancer therapy. METHODOLOGY/PRINCIPAL FINDINGS:We describe the initial events of colonization of an ectopic transplantable tumor by Salmonella enterica serovar Typhimurium. Initially, after intravenous administration, bacteria were found in blood, spleen, and liver. Low numbers were also detected in tumors associated with blood vessels as could be observed by immunohistochemistry. A rapid increase of TNF-alpha in blood was observed at that time, in addition to other pro-inflammatory cytokines. This induced a tremendous influx of blood into the tumors by vascular disruption that could be visualized in H&E stainings and quantified by hemoglobin measurements of tumor homogenate. Most likely, together with the blood, bacteria were flushed into the tumor. In addition, blood influx was followed by necrosis formation, bacterial growth, and infiltration of neutrophilic granulocytes. Depletion of TNF-alpha retarded blood influx and delayed bacterial tumor-colonization. CONCLUSION:Our findings emphasize similarities between Gram-negative tumor-colonizing bacteria and tumor vascular disrupting agents and show the involvement of TNF-alpha in the initial phase of tumor-colonization by bacteria

    Retinoic acid induces homing of protective T and B cells to the gut after subcutaneous immunization in mice

    No full text
    Diarrheal diseases represent a major health burden in developing countries. Parenteral immunization typically does not induce efficient protection against enteropathogens because it does not stimulate migration of immune cells to the gut. Retinoic acid (RA) is critical for gut immunity, inducing upregulation of gut-homing receptors on activated T cells. In this study, we have demonstrated that RA can redirect immune responses elicited by s.c. vaccination of mice from skin-draining inguinal LNs (ingLNs) to the gut. When present during priming, RA induced robust upregulation of gut-homing receptors in ingLNs, imprinting gut-homing capacity on T cells. Concurrently, RA triggered the generation of gut-tropic IgA+ plasma cells in ingLNs and raised the levels of antigen-specific IgA in the intestinal lumen and blood. RA applied s.c. in vivo induced autonomous RA production in ingLN DCs, further driving efficient induction of gut-homing molecules on effector cells. Importantly, RA-supplemented s.c. immunization elicited a potent immune response in the small intestine that protected mice from cholera toxin–induced diarrhea and diminished bacterial loads in Peyer patches after oral infection with Salmonella. Thus, the use of RA as a gut-homing navigator represents a powerful tool to induce protective immunity in the intestine after s.c. immunization, offering what we believe to be a novel approach for vaccination against enteropathogens

    TNF-α release into the blood of <i>S.</i> Typhimurium-infected, CT26 tumor-bearing mice.

    No full text
    <p>(a) RT-PCRs were performed with RNA isolated from <i>S.</i> Typhimurium-infected CT26 cells (<i>in vitro</i>) and from CT26 tumors of <i>S.</i> Typhimurium-infected, tumor-bearing BALB/c mice (<i>in vivo</i>) at the indicated time points. The amplification product has a size of 212 bp. (b) TNF-α concentration in the blood of CT26 tumor-bearing BALB/c mice at different times p.i. determined using TNF sensitive L929 fibroblasts. Error bars show standard deviations of means. Results are representative for at least two independent experiments with 3–5 mice per group.</p

    Inhibition of TNF-α in <i>S.</i> Typhimurium-infected BALB/c mice retards blood influx into tumors and bacterial colonization.

    No full text
    <p>(a) TNF-α concentration in the blood of <i>S.</i> Typhimurium-infected, CT26 tumor-bearing BALB/c mice (black bars) and of <i>S.</i> Typhimurium-infected, anti-TNF-α treated, CT26 tumor-bearing BALB/c mice (grey bars). Error bars show standard deviations of means. (b) Photographs of CT26 tumors of an <i>S.</i> Typhimurium-infected tumor (left set of pictures) and an <i>S.</i> Typhimurium-infected, anti-TNF-α treated tumor (right set of pictures). The left column of each set of pictures show the fur side of the tumors, the right pictures show the ventral side of the tumors. (c) Hemoglobin content in CT26 tumors of <i>S.</i> Typhimurium-infected BALB/c mice (black bars) and <i>S.</i> Typhimurium-infected, anti-TNF-α treated BALB/c mice (grey bars). Error bars show standard deviations of means. (*) At 6 h p.i. the difference of hemoglobin content in the tumors is significant with p<0.05. (d) Bacterial number per g tumor tissue in <i>S.</i> Typhimurium-infected tumor-bearing BALB/c mice (black bars) and <i>S.</i> Typhimurium-infected, anti-TNF-α treated tumor-bearing BALB/c mice (grey bars). Error bars show standard deviations of means. (*) At 12 h p.i. the difference of bacterial numbers between differently treated tumors is significant with p<0.05. Results are representative for at least two independent experiments with 3–5 mice per group. The second experiment also included an isotype control where normal rat IgG was injected into the mice of the control group (data not shown).</p

    Time course of blood influx into CT26 tumors after bacterial infection.

    No full text
    <p>(a) Photographs of CT26 tumors at different time points p.i. with <i>S.</i> Typhimurium. (a left) Photographs of the fur side of the tumors. (a right) Photographs of the ventral side of the tumors. (b) HE-stained paraffin sections of CT26 tumors at different time points p.i. with <i>S.</i> Typhimurium. (b left) Low magnification overviews. (b right) Higher magnifications of the tumors shown in (b left). 6 h–12 h: High magnifications of erythrocyte-containing (stars) central tumor areas. 15 h–24 h: High magnifications of the transition between developing necrosis (N) and remaining vital tumor rim (V). Arrows point exemplarily at neutrophils. Black bars correspond to 500 µm in (b left) and 100 µm in (b right). (c) Hemoglobin content in CT26 tumors at different time points p.i. with <i>S.</i> Typhimurium. Bars show standard deviations of means. Experiments were repeated three times with identical results.</p

    Time course of bacterial accumulation in different organs and bacterial colonization of solid CT26 tumors.

    No full text
    <p>Tumor bearing mice were infected i.v. with <i>S.</i> Typhimurium SL7207. (a) 2 h, 6 h, 12 h, 24 h and 48 h p.i. tumor and spleen were homogenized. Tumor, spleen and blood were plated and the CFUs per total organ were determined. (b) Non-invasive <i>in vivo</i> imaging of bacterial bioluminescence. Tumor-bearing mice were infected with <i>S.</i> Typhimurium expressing the luxCDABE operon of <i>Photorhabdus luminescens</i> under the control of the β-Lactamase promoter. Arrows show sites of high bacterial accumulation. Images were taken at the indicated time points. (c) High magnification images of tumor cryosections at the indicated time points p.i. with <i>S.</i> Typhimurium SL7207. In all pictures, bacteria are stained in green, blood vessel endothelial cells are stained in blue and neutrophilic granulocytes are stained in red. White arrows point at individual Salmonella. White bars correspond to 10 µm in all pictures. (d) Overviews of CT26 tumor cryosections at the indicated time points p.i. with <i>S.</i> Typhimurium SL7207. Stainings are described in (c). White bars correspond to 100 µm in all pictures. Experiments were repeated at least three times with identical results.</p

    Measurement of different proinflammatory cytokines in the initial phase of bacterial tumor colonization.

    No full text
    <p>Concentrations of the proinflammatory cytokines TNF-α, IL-6, MCP-1, IFN-γ, IL-10 and IL-12(p70) in blood of uninfected (0 h) and <i>S.</i> Typhimurium-infected (2 h, 6 h) CT26 tumor-bearing BALB/c mice at different time points p.i.. Error bars show standard deviations. The differences between <i>S.</i> Typhimurium-infected mice and uninfected control mice are significant with p<0.01–0.05. The 6 h-values for IL-10 and IL-12 are not significantly different from non-infected mice. According to the manufacturer's protocol, the values for IL-12 can be considered background. The exact values for MCP-1 are higher than plotted here since several samples reached the plateau. Results are representative for two independent experiments with 3–5 mice.</p
    corecore