Peanut Induced Dendritic Cell (DC) Maturation and Cytokine Production Is Modulated by the Microbial Components of EMP-123, Leading to Changes in Adaptive Immunity In Vitro

2009 ◽  
Vol 123 (2) ◽  
pp. S211-S211
Author(s):  
B.P. Vickery ◽  
P. Pochard ◽  
M.J. Caplan ◽  
H.A. Sampson ◽  
M.C. Berin ◽  
...  
Author(s):  
Anja Ziegler ◽  
Judith Olzhausen ◽  
Eman Hamza ◽  
Ana Stojiljkovic ◽  
Michael H. Stoffel ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (9) ◽  
pp. 2473-2482 ◽  
Author(s):  
Catharina H. M. J. Van Elssen ◽  
Joris Vanderlocht ◽  
Tammy Oth ◽  
Birgit L. M. G. Senden-Gijsbers ◽  
Wilfred T. V. Germeraad ◽  
...  

Abstract Among prostaglandins (PGs), PGE2 is abundantly expressed in various malignancies and is probably one of many factors promoting tumor growth by inhibiting tumor immune surveillance. In the current study, we report on a novel mechanism by which PGE2 inhibits in vitro natural killer–dendritic cell (NK-DC) crosstalk and thereby innate and adaptive immune responses via its effect on NK-DC crosstalk. The presence of PGE2 during IFN-γ/membrane fraction of Klebsiella pneumoniae DC maturation inhibits the production of chemokines (CCL5, CCL19, and CXCL10) and cytokines (IL-12 and IL-18), which is cAMP-dependent and imprinted during DC maturation. As a consequence, these DCs fail to attract NK cells and show a decreased capacity to trigger NK cell IFN-γ production, which in turn leads to reduced T-helper 1 polarization. In addition, the presence of PGE2 during DC maturation impairs DC-mediated augmentation of NK-cell cytotoxicity. Opposed to their inhibitory effects on peripheral blood–derived NK cells, PGE2 matured DCs induce IL-22 secretion of inflammation constraining NKp44+ NK cells present in mucosa-associated lymphoid tissue. The inhibition of NK-DC interaction is a novel regulatory property of PGE2 that is of possible relevance in dampening immune responses in vivo.


Blood ◽  
2006 ◽  
Vol 107 (4) ◽  
pp. 1459-1467 ◽  
Author(s):  
Chang-Hung Chen ◽  
Helen Floyd ◽  
N. Eric Olson ◽  
Dario Magaletti ◽  
Chang Li ◽  
...  

Dendritic-cell (DC)-associated C-type lectin receptors (CLRs) take up antigens to present to T cells and regulate DC functions. DCAL-2 is a CLR with a cytosolic immunoreceptor tyrosine-based inhibitory motif (ITIM), which is restricted to immature DCs (iDCs), monocytes, and CD1a+ DCs. Cross-linking DCAL-2 on iDCs induced protein tyrosine phosphorylation and MAPK activation as well as receptor internalization. To test if DCAL-2 is involved in DC maturation and cytokine expression, we stimulated iDCs with anti-DCAL-2 mAb with or without LPS, zymosan, or CD40L. While anti-DCAL-2 did not induce iDCs to mature, it did up-regulate CCR7 expression and IL-6 and IL-10 production. DCAL-2 signals augmented DC maturation induced by LPS or zymosan, increasing both CCR7 and DC-LAMP expression. Of interest, DCAL-2 ligation had the opposite effects on TLR versus CD40L signaling: anti-DCAL-2 suppressed TLR-induced IL-12 expression, but significantly enhanced CD40L-induced IL-12 production. DCAL-2 ligation also suppressed the ability of TLR-matured DCs to induce IFN-γ-secreting Th1 cells but augmented the capacity of CD40L-matured DCs to polarize naive T cells into Th1 cells. Thus, DCAL-2 may program DCs differently depending on whether DCs are signaled via TLRs or by T cells. DCAL-2 may be a potential immunotherapeutic target for modulating autoimmune diseases or for developing vaccines.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4118-4118
Author(s):  
Haein Park ◽  
Xin Huang ◽  
Changming Lu ◽  
Mitchell S. Cairo ◽  
Xianzheng Zhou

Abstract MicroRNA (miRNA) regulation of dendritic cell (DC) development and function is not fully understood. We have previously reported 27 differentially expressed miRNAs during human monocyte differentiation into immature DCs (imDCs) and mature DCs (mDCs). Here, we aimed at uncovering the functional role of miR-146a and miR-146b (miR-146a/b) during this differentiation process. To investigate miR-146a and miR-146b expression during human monocyte differentiation into imDCs and mDCs, monocytes were differentiated into imDCs with GM-CSF and IL-4 and matured with IL-1β, IL-6, TNF-α, and PGE2. We found by qRT-PCR that expression of miR-146a/b was dramatically increased upon monocyte differentiation into imDCs (miR-146a, 10-fold; miR-146b, 37-fold at day6; n=4; p value of Student t test<0.05) and mDCs (miR-146a, 51-fold; miR-146b, 79-fold; n=4; p<0.005). Up-regulation of miR-146a/b in mDCs was predominantly mediated by IL-1β but not IL-6, TNF-a, or PGE2 (n=3; p<0.01). DC apoptosis is important for self-tolerance and immunity. We then evaluated the effect of altering miR-146a/b expression levels on DC apoptosis by Annexin V/PI staining. Silencing of miR-146a, miR-146b or both in imDCs (Fig. 1A) and mDCs significantly prevented DC from apoptosis (Fig. 1A; miR-146a, 24%±3.1; miR-146b, 26%±1.7; miR-146a/b; 23%±2.8 vs control, 46%±2.3 of Annexin V+ populations; n=8; p<0.005) whereas overexpression of miR-146a, miR-146b or both in imDCs (Fig. 1B) and mDCs significantly increased the proportion of apoptotic cells (Fig. 1B; 46%±1.9, 47%±2.1, 45%±2.3 vs 36%±3.3; n=6; p<0.05). These results indicate that miR-146a/b may function as pro-apoptotic regulators during human monocyte differentiation into imDCs and mDCs. It is known that the NF-κB pathway regulates DC development, function and survival, and that TRAF6 and IRAK1 are major signal transducers in the NF-κB pathway. In addition, both TRAF6 and IRAK1 are known target genes of miR-146a. Indeed, miR-146a/b expression in imDCs and mDCs was inversely correlated with TRAF6 and IRAK1 mRNA and protein expression (n=4; p<0.05). Furthermore, siRNA silencing of TRAF6 and/or IRAK1 in imDCs and mDCs significantly enhanced DC apoptosis (n=4; p<0.05). By contrast, lentivirus overexpression of TRAF6 and/or IRAK1 promoted DC survival compared to control lentivirus transduced cells (n=2; p<0.05). To confirm that miR-146a/b-induced human DC apoptosis is involved in suppression of the NF-κB pathway, at least in part through down regulation of the NF-κB signaling transducers TRAF6 and IRAK1, we examined the protein level of IκB as a negative regulator of NF-κB and Bcl-2 as a known downstream anti-apoptotic molecule of the NF-κB pathway. Silencing of miR-146a/b in imDCs and mDCs significantly decreased IκBα and increased Bcl-2 expression whereas overexpression of miR-146a and/or miR-146b or silencing of TRAF6 and/or IRAK1 significantly increased IκBα and decreased Bcl-2 expression in imDCs and mDCs (n=4; p<0.01). These results indicate that miR-146a/b modulate DC apoptosis through inhibition of NF-κB activation via targeting TRAF6 and IRAK1. Next, we investigated whether miR-146a/b regulates pro-inflammatory cytokine production in DCs. We found that IL-12p70, IL-6 and TNF-α production were significantly enhanced after miR-146 and/or miR-146b silencing during DC maturation (n≥2; p<0.05), although altering miR-146a/b expression had little effect on DC maturation (n=6). By contrast, IL-12p70, IL-6 and TNF-α production was highly reduced after miR-146a and/or miR-146b overexpression (n≥2; p<0.05). In conclusion, we have demonstrated three important findings in this report. First, expression of both miR-146a and miR-146b is up-regulated during human monocyte differentiation into imDCs and mDCs. Secondly, although miR-146a and miR-146b do not appear to play a role in DC maturation, they may be critical regulators of DC apoptosis and cytokine production. Thirdly, mechanistically, miR-146a/b targets TRAF6 and IRAK1, leading to inhibition of NF-κB and reduced expression of Bcl-2. We thus demonstrate for the first time that miR-146a/b regulates human DC apoptosis and cytokine production, uncovering a new negative feedback mechanism for miR-146 in controlling overstimulation of the immune responses (Fig. 2). Disclosures No relevant conflicts of interest to declare.


2016 ◽  
Vol 22 (5) ◽  
pp. 316-324 ◽  
Author(s):  
Fabio SY Yoshikawa ◽  
Rikio Yabe ◽  
Yoichiro Iwakura ◽  
Sandro R de Almeida ◽  
Shinobu Saijo

Dermatophytoses are chronic fungal infections, the main causative agent of which is Trichophyton rubrum (T. rubrum). Despite their high occurrence worldwide, the immunological mechanisms underlying these diseases remain largely unknown. Here, we uncovered the C-type lectin receptors, Dectin-1 and Dectin-2, as key elements in the immune response to T. rubrum infection in a model of deep dermatophytosis . In vitro, we observed that deficiency in Dectin-1 and Dectin-2 severely compromised cytokine production by dendritic cells. In vivo, mice lacking Dectin-1 and/or Dectin-2 showed an inadequate pro-inflammatory cytokine production in response to T. rubrum infection, impairing its resolution. Strikingly, neither adaptive immunity nor IL-17 response were required for fungal clearance, highlighting innate immunity as the main checkpoint in the pathogenesis of T. rubrum infection.


Sign in / Sign up

Export Citation Format

Share Document