Fig 3 - uploaded by Janosch Klebensberger
Content may be subject to copyright.
PGE 2 induces active IDO protein, 

PGE 2 induces active IDO protein, 

Source publication
Article
Full-text available
The exclusive ability of dendritic cells (DCs) to stimulate primary and secondary immune responses favors the use of antigen-loaded human monocyte-derived DCs (MoDCs) in vaccinations against tumors. Previous studies demonstrated that PGE(2) is fundamental during MoDC maturation to facilitate migration toward lymph node-derived chemokines. A recent...

Contexts in source publication

Context 1
... or absence of PGE 2 for 2 days, and mature cells were incubated with tryptophan. To determine tryptophan degradation, we quantified residual tryptophan concentration as well as gener- ated kynurenine in culture supernatants by HPLC. As ex- pected, MoDCs matured with sCD40L alone, showing unde- tectable IDO expression, did not degrade tryptophan (Fig. 3C). In contrast, IDO from the supernatant of MoDCs matured with sCD40L and PGE 2 readily degraded tryptophan to kynurenine (Fig. 3C). Correlating with IDO expression, MoDCs matured with TLR3 ligand poly(I:C) produced active IDO indepen- dently of PGE 2 , as more than 70% of tryptophan was converted to kynurenine in the presence as well as ...
Context 2
... residual tryptophan concentration as well as gener- ated kynurenine in culture supernatants by HPLC. As ex- pected, MoDCs matured with sCD40L alone, showing unde- tectable IDO expression, did not degrade tryptophan (Fig. 3C). In contrast, IDO from the supernatant of MoDCs matured with sCD40L and PGE 2 readily degraded tryptophan to kynurenine (Fig. 3C). Correlating with IDO expression, MoDCs matured with TLR3 ligand poly(I:C) produced active IDO indepen- dently of PGE 2 , as more than 70% of tryptophan was converted to kynurenine in the presence as well as absence of PGE 2 during the maturation process (Fig. ...
Context 3
... of MoDCs matured with sCD40L and PGE 2 readily degraded tryptophan to kynurenine (Fig. 3C). Correlating with IDO expression, MoDCs matured with TLR3 ligand poly(I:C) produced active IDO indepen- dently of PGE 2 , as more than 70% of tryptophan was converted to kynurenine in the presence as well as absence of PGE 2 during the maturation process (Fig. ...
Context 4
... for clini- cal trials, we matured MoDCs with sCD40L in the absence or presence of PGE 2 or two specific EP4 receptor agonists, ONO- AE1-329 and PGE 1 alcohol. It is surprising that under these conditions, IDO protein was induced and fully active if MoDCs were matured in the presence of either of the two EP4 agonists and were comparable with PGE 2 (Figs. 3C and 4A). It is unexpected that EP4 receptor-induced IDO activity was even higher than EP2 receptor-mediated IDO induction by butaprost (Fig. 3D). The ineffective degradation of tryptophan by MoDCs matured with sCD40L in the presence of butaprost correlated with low induction of IDO protein expression (Fig. 4A). To ensure functionality of the ...
Context 5
... 1 alcohol. It is surprising that under these conditions, IDO protein was induced and fully active if MoDCs were matured in the presence of either of the two EP4 agonists and were comparable with PGE 2 (Figs. 3C and 4A). It is unexpected that EP4 receptor-induced IDO activity was even higher than EP2 receptor-mediated IDO induction by butaprost (Fig. 3D). The ineffective degradation of tryptophan by MoDCs matured with sCD40L in the presence of butaprost correlated with low induction of IDO protein expression (Fig. 4A). To ensure functionality of the EP2 agonist, we analyzed the mi- gratory behavior of MoDCs. We matured MoDCs with sCD40L in the absence or presence of PGE 2 , the EP2 ...
Context 6
... maturation provoked up-regulation of IDO on mRNA and protein level (Figs. 1 and 2A), confirming and extending the observations by Braun et al. [20]. However, we demonstrated that TLR3-medi- ated MoDC maturation using poly(I:C) induced IDO expres- sion, independently of PGE 2 (Figs. 1 and 2A), and poly(I:C)- induced IDO protein was fully active (Fig. 3B). In our hands, IDO protein expression strictly correlated with the enzyme's activity. Thus, PGE 2 is not a general prerequisite for IDO expression in mature MoDCs, as implied previously ...
Context 7
... of active IDO. In striking contrast, using two independent, specific agonists, we clearly demonstrate that under serum-free, clinically rele- vant conditions, PGE 2 -induced IDO protein expression and activity are mediated primarily by the PGE 2 receptor EP4 (Figs. 3C and 4A). EP2 triggering also induced IDO activity but to a much lower level (Fig. 3D), which correlated with lower IDO protein induction (Fig. 4A). The addition of PGE 2 , EP2, or EP4 agonists during MoDC maturation is not only essential for the development of a migratory phenotype but also induces active IDO protein. It is interesting that IDO may even be critical for DC activation and chemotaxis, as incubation of ...

Citations

... In addition to this modulation toward immune suppression, PGE2 also confers proinflammatory DC characteristics, including the upregulation of co-stimulatory markers (e.g. CD40, CD80, and CD86) and the antigen-presenting molecule MHC class II, leading to a superior capacity to stimulate and expand T cells [15,20,[23][24][25][26]. Furthermore, it is well-known that PGE2 upregulates CCR7 facilitating DC migration toward lymph nodes [27][28][29][30]. ...
... Expansion of T cells is highly dependent on the full maturation state of cDC2s [43]. Like what was previously reported for moDCs [24,25], cDC2s treated with MC+PGE2 exhibited a superior T-cell expansion capacity as compared with cDC2s matured in the absence of PGE2. This superior expansion was to be expected, as T-cell expansion is mostly dependent on the expression of co-stimulatory molecules by DCs. ...
Article
Full-text available
Dendritic cells (DCs) shape adaptive immunity in response to environmental cues such as cytokines or lipid mediators, including prostaglandin E2 (PGE2). In cancer, tumors are known to establish an enriched PGE2‐microenvironment. Tumor‐derived PGE2 primes regulatory features across immune cells, including DCs, facilitating tumor progression. PGE2 shapes DC function by providing signaling via its two so‐called E‐prostanoid receptors (EPs) EP2 and EP4. While studies with monocyte‐derived DCs (moDCs) have shown the importance of PGE2 signaling, the role PGE2‐EP2/EP4 on conventional DCs type 2 (cDC2s), is still poorly defined. In this study, we investigated the function of EP2 and EP4 using specific EP antagonists on human cDC2s. Our results show that EP2 and EP4 exhibit different functions in cDC2s, with EP4 modulating the upregulation of activation markers (CD80, CD86, CD83, MHC class II) and the production of interleukin 10 (IL‐10) and IL‐23. Furthermore, PGE2‐EP4 boosts CCR type 7 (CCR7) based migration as well as a higher T cell expansion capacity, characterized by the enrichment of suppressive rather than pro‐inflammatory T cell populations. Our findings are relevant to further understanding the role of EP receptors in cDC2s, underscoring the benefit of targeting PGE2‐EP2/4 axis for therapeutic purposes in diseases such as cancer. This article is protected by copyright. All rights reserved
... However, this pro-regulatory capacity did not seem to be sufficient to overcome T cell stimulation, probably due to the counteraction of all the costimulatory molecules they contain and the absence of inhibitory signals. This feature has been well described by others who have discussed the ambivalent and paradoxical relevance of IDO expression by mature DC, particularly in cancer therapy (Harden & Egilmez, 2012;Krause et al., 2007). On the contrary, NPCSEV-DCs showed impaired production of proinflammatory cytokines IL-6 and IL-12. ...
Article
Full-text available
Nasopharyngeal carcinoma‐derived small extracellular vesicles (NPCSEVs) have an immunosuppressive impact on the tumour microenvironment. In this study, we investigated their influence on the generation of tolerogenic dendritic cells and the potential involvement of the galectin‐9 (Gal9) they carry in this process. We analysed the phenotype and immunosuppressive properties of NPCSEVs and explored the ability of DCs exposed to NPCSEVs (NPCSEV‐DCs) to regulate T cell proliferation. To assess their impact at the pathophysiological level, we performed real‐time fluorescent chemoattraction assays. Finally, we analysed phenotype and immunosuppressive functions of NPCSEV‐DCs using a proprietary anti‐Gal9 neutralising antibody to assess the role of Gal9 in this effect. We described that NPCSEV‐DCs were able to inhibit T cell proliferation despite their mature phenotype. These mature regulatory DCs (mregDCs) have a specific oxidative metabolism and secrete high levels of IL‐4. Chemoattraction assays revealed that NPCSEVs could preferentially recruit NPCSEV‐DCs. Finally, and very interestingly, the reduction of the immunosuppressive function of NPCSEV‐DCs using an anti‐Gal9 antibody clearly suggested an important role for vesicular Gal9 in the induction of mregDCs. These results revealed for the first time that NPCSEVs promote the emergence of mregDCs using a galectin‐9 dependent mechanism and open new perspectives for antitumour immunotherapy targeting NPCSEVs.
... В целом, стимуляция созревания незрелых ДК сводится к 2 основным способам: (1) воздействие провоспалительных цитокинов и (2) использование лигандов для патоген-распознающих рецепторов (PRR), а также их смесей [9]. В данной работе нами была оценена стимулирующая активность TNFα и цитокинового «коктейля» из TNFα, IL-1, IL-6 с добавлением PGE2 [23]. Также были использованы лиганды PRR: poly I:C, имитирующий вирусную 2-цепочечную РНК (лиганд TLR3), LPS (лиганд TLR4) и, кроме того, лизированные клетки меланомы как источник DAMP (danger-associated molecular patterns, «молекулярные образы, ассоциированные с опасностью»), которые также являются триггерами для созревания ДК [14]. ...
Article
Monocyte-derived dendritic cells (DCs) can be used for cell immunotherapy of cancer. In most cases, mature DCs, loaded with tumor-associated antigens, are used for immune therapy. The functionality of DCs for immunotherapy substantially depends on their immunophenotype and secretory profile, which are established after DCs maturation. The purpose of this research was to explore the phenotype of DCs after using various approaches for stimulation of their maturation. Maturation of DCs was stimulated by pro-inflammatory cytokines and their mixtures, or by ligands to the TLRs of DCs. DCs were stimulated by the following means: TNF; poly I:C; LPS; cytokine cocktail (TNF + IL-1 + IL-6 + PGE2); the cocktail mixed with poly I:C; and melanoma cells lysate. Forty-eight hours after stimulation, the expression of DCs’ receptors involved into their interaction with T cells, was evaluated by flow cytometry. Moreover, the secretion of IL-12 (activator of T cell response) and IL-10 (inhibitor of T cell response) was estimated by ELISA technique. We have shown that, following stimulation with cytokine cocktail, the DCs exhibit highest expression of receptors, which are necessary for interaction with T cells and for activation of T cell mediated immune response, i.e., antigen-presenting receptors (HLA-DR), co-stimulatory receptors (CD83, CD40, CD86), and receptors controlling the migration of DCs to lymph nodes (CCR7). Moreover, the cocktail-stimulated DCs intensively secrete both IL-12 and IL-10. The stimulatory effect of TNF and poly I:C proved to be moderate: the expression of most receptors was significantly lower than after using the cocktail; no significant differences from control (in absence of induced maturation) in IL-12 secretion were detected. LPS and melanoma cell lysate did not affect both expression of receptors and secretory profile of DCs. Addition of poly I:C to the cytokine cocktail did not affect the receptor expression, but significantly increased the secretion of both proinflammatory IL-12 and anti-inflammatory IL-10. The results of experiments demonstrate that the mixture of cytokine cocktail and poly I:C seems to be the most effective tool for stimulation of DCs maturation. However, further experiments are required to compare the functionality of DCs when using different tools for induced DC maturation.
... This study also indicated that I/R AKI mice treated with prostaglandin E2 (PGE2, 0.5 mg/mL administered daily interperitoneally for 14 days) generated similar results to the IDO1 deficient mice. PGE2 is also known to induce the transcription and production of IDO1 [107], highlighting the complexity of this response that may depend on the select activation of one of the four PGE2 receptors [108]. ...
Article
Full-text available
Early-stage detection of chronic kidney diseases (CKD) is important to treatment that may slow and occasionally halt CKD progression. CKD of diverse etiologies share similar histologic patterns of glomerulosclerosis, tubular atrophy, and interstitial fibrosis. Macro-vascular disease and micro-vascular disease promote tissue ischemia, contributing to injury. Tissue ischemia promotes hypoxia, and this in turn activates the hypoxia-inducible transcription factors (HIFs). HIF-1α and HIF-2α, share a dimer partner, HIF-1β, with the aryl hydrocarbon receptor (AHR) and are each activated in CKD and associated with kidney cellular nicotinamide adenine dinucleotide (NAD) depletion. The Preiss-Handler, salvage, and de novo pathways regulate NAD biosynthesis and gap-junctions regulate NAD cellular retention. In the Preiss-Handler pathway, niacin forms NAD. Niacin also exhibits crosstalk with HIF and AHR cell signals in the regulation of insulin sensitivity, which is a complication in CKD. Dysregulated enzyme activity in the NAD de novo pathway increases the levels of circulating tryptophan metabolites that activate AHR, resulting in poly-ADP ribose polymerase activation, thrombosis, endothelial dysfunction, and immunosuppression. Therapeutically, metabolites from the NAD salvage pathway increase NAD production and subsequent sirtuin deacetylase activity, resulting in reduced activation of retinoic acid-inducible gene I, p53, NF-κB and SMAD2 but increased activation of FOXO1, PGC-1α, and DNA methyltransferase-1. These post-translational responses may also be initiated through non-coding RNAs (ncRNAs), which are additionally altered in CKD. Nanoparticles traverse biological systems and can penetrate almost all tissues as disease biomarkers and drug delivery carriers. Targeted delivery of non-coding RNAs or NAD metabolites with nanoparticles may enable the development of more effective diagnostics and therapies to treat CKD.
... Human DC subsets can be isolated in very limited numbers from peripheral blood (11) and hence are not exploited as cellular model systems to study molecular mechanisms of DC migration. The most widely used human DC model system is monocyte-derived DCs (MoDCs), which comprises the isolation of peripheral blood monocytes and their in vitro differentiation to MoDCs in the presence of GM-CSF and IL-4 for several days (7,(25)(26)(27)(28). A drawback of these MoDCs is that they show substantial donor to donor variation, are refractory to genetic manipulation and hence not suitable as effective cellular model to study CCR7-driven DC migration. ...
Article
Full-text available
Dendritic cells (DCs) are potent and versatile professional antigen-presenting cells and central for the induction of adaptive immunity. The ability to migrate and transport peripherally acquired antigens to draining lymph nodes for subsequent cognate T cell priming is a key feature of DCs. Consequently, DC-based immunotherapies are used to elicit tumor-antigen specific T cell responses in cancer patients. Understanding chemokine-guided DC migration is critical to explore DCs as cellular vaccines for immunotherapeutic approaches. Currently, research is hampered by the lack of appropriate human cellular model systems to effectively study spatio-temporal signaling and CCR7-driven migration of human DCs. Here, we report that the previously established human neoplastic cell line CAL-1 expresses the human DC surface antigens CD11c and HLA-DR together with co-stimulatory molecules. Importantly, if exposed for three days to GM-CSF, CAL-1 cells induce the endogenous expression of the chemokine receptor CCR7 upon encountering the clinically approved TLR7/8 agonist Resiquimod R848 and readily migrate along chemokine gradients. Further, we demonstrate that CAL-1 cells can be genetically modified to express fluorescent (GFP)-tagged reporter proteins to study and visualize signaling or can be gene-edited using CRISPR/Cas9. Hence, we herein present the human CAL-1 cell line as versatile and valuable cellular model system to effectively study human DC migration and signaling.
... Furthermore, PGE2/EP4 signaling plays critical roles in stem cell expansion, metastasis, and tumor microenvironment of CRC [10,11]. For instance, the expression of indoleamine 2,3-dioxygenase (IDO), an enzyme that plays a role in induction of immune tolerance, is regulated by PGE2/EP4 signaling [12,13]. Consequently, targeting PGE2/EP4 signaling can be a promising approach for the prevention and treatment of CRC [14,15]. ...
... RQ-15986 attenuated the inflammation in colonic mucosa, which might lead to IDO suppression. In CRC tissues, IDO might be suppressed by RQ-15986 in the similar way as previously reported that inhibition of EP4 downregulated IDO expression in cancer cells and monocyte-derived dendritic cells [12,13]. According to the previous reports [7,13], and the present study, both EP4 and IDO appear positive on the epithelial cells and tumor cells, and both EP4 and IDO are presumably thought to express in the same cells. ...
Article
Full-text available
Prostaglandin E2 receptor EP4 is involved in inflammation and related tumorigenesis in the colorectum. This study aimed to investigate the chemopreventive ability of RQ-15986, a selective EP4 antagonist, in colitis-related colorectal tumorigenesis. Male Kyoto APC delta rats, which have APC mutations, were treated with azoxymethane and dextran sulfate sodium and subsequently administered RQ-15986 for eight weeks. At the end of the experiment, the development of colorectal tumor was significantly inhibited in the RQ-15986-treated group. The cell proliferation of the crypts and tumors in the colorectum was decreased following RQ-15986 treatment. RQ-15986 also suppressed the expression of pro-inflammatory cytokines, including tumor necrosis factor-α, interleukin-6, interleukin-18, and monocyte chemotactic protein-1, in the colon mucosa. In addition, the expression levels of indoleamine 2,3-dioxygenase, which is involved in immune tolerance, were decreased in the colorectal epithelium and tumors of the RQ-15986-treated group. These findings indicate that RQ-15986 inhibits colitis-associated colorectal tumorigenesis by attenuating inflammation, suppressing cell proliferation, and modulating the expression of indoleamine 2,3-dioxygenase. Targeting prostaglandin E2/EP4 signaling might be a useful strategy for chemoprevention of inflammation-related colorectal cancer.
... However, due to the effects of MSCs, immature DC precursors (CD34 + precursors) ceased at the differentiation [72][73][74]. PGE2 from MSCs has been reported for its tolerogenic features, such as IL-10 release, IDO1 expression, also when merged with TNF-α, IL-1β, and IL6, it intensifies the immunogenicity with the co-stimulatory molecules [75][76][77]. In the vicinity of microenvironments treated with MSCs, mature DCs (CD83 + DCs with CD80 and CD86 costimulatory molecules) get obstructed in their efficiency for T cell activation [74,78]. ...
Article
Full-text available
The pleiotropic behavior of mesenchymal stem cells (MSCs) has gained global attention due to their immense potential for immunosuppression and their therapeutic role in immune disorders. MSCs migrate towards inflamed microenvironments, produce anti-inflammatory cytokines and conceal themselves from the innate immune system. These signatures are the reason for the uprising in the sciences of cellular therapy in the last decades. Irrespective of their therapeutic role in immune disorders, some factors limit beneficial effects such as inconsistency of cell characteristics, erratic protocols, deviating dosages, and diverse transfusion patterns. Conclusive protocols for cell culture, differentiation, expansion, and cryopreservation of MSCs are of the utmost importance for a better understanding of MSCs in therapeutic applications. In this review, we address the immunomodulatory properties and immunosuppressive actions of MSCs. Also, we sum up the results of the enhancement, utilization, and therapeutic responses of MSCs in treating inflammatory diseases, metabolic disorders and diabetes.
... PGE2 inhibits dendritic cell maturation, activation and antigen presentation to prevent MSCs from being recognized as foreign bodies. 25 IDO inhibits B-cell proliferation by catalyzing the conversion of tryptophan to kynurenine. 22 MSCs can be primed by the activated immune cells to boost the antiinflammatory properties. ...
... Furthermore, we could show that the anti-leukemic activity is comparable and it might be superior after MLC DC/DC leu generated with the PGE 1 -containing Kit M when compared to the PGE 2 -containing Kit K, especially after 24 h of simultaneous incubation of the effector and target cells. This finding might be explained by the fact that PGE 2 could induce the expression of indoleamin 2,3-dioxgenase-1 (IDO1), an immunoregulatory enzyme that activates immunosuppressive T reg , but does not impair the antigen presentation capacity of DCs [70][71][72][73][74]. ...
Article
Full-text available
Dendritic cells (DCs) and leukemia-derived DC (DCleu) are potent stimulators of various immunoreactive cells and they play a pivotal role in the (re-) activation of the immune system. As a potential treatment tool for patients with acute myeloid leukemia, we developed and analyzed two new PGE1-containing protocols (Pici-PGE1, Kit M) to generate DC/DCleu ex vivo from leukemic peripheral blood mononuclear cells (PBMCs) or directly from leukemic whole blood (WB) to simulate physiological conditions. Pici-PGE1 generated significantly higher amounts of DCs from leukemic and healthy PBMCs when compared to control and comparable amounts as the already established protocol Pici-PGE2. The proportions of sufficient DC-generation were even higher after DC/DCleu-generation with Pici-PGE1. With Kits, it was possible to generate DCs and DCleu directly from leukemic and healthy WB without induction of blast proliferation. The average amounts of generated DCs and DCleu-subgroups were comparable with all Kits. The PGE1 containing Kit M generated significantly higher amounts of mature DCs when compared to the PGE2-containing Kit K and increased the anti-leukemic-activity. In summary PGE1-containing protocols were suitable for generating DC/DCleu from PBMCs as well as from WB, which reliably (re-) activated immunoreactive cells, improved the overall ex vivo anti-leukemic activity, and influenced cytokine-release-profiles.
... In the earlier trials, the gold standard maturation cocktail included the pro-inflammatory cytokines TNF-α, IL-1β, and IL-6 in combination with prostaglandin E2 (PGE2) (8)(9)(10). However, despite the important roles of PGE2 in promoting DC migration (11) and in enhancing T cell proliferation (12), it has also been shown that PGE2 may induce differentiation of regulatory T cells (13), increase the expression of the pro-tolerogenic enzyme indoleamine 2,3-dioxygenase (IDO) (14), and may limit IL-12p70 production (15). As these PGE2-related activity may curtail the anti-tumoral immune response, alternative methods of ex vivo maturation of DC have been explored such as the triggering of co-stimulatory pathways (e.g., CD40-CD40L) (16) and the activation of the TLR using agonists such as poly IC (TLR3) (17), resiquimod (TLR7/8) (8) and 3-O-deacylated monophosphoryl lipid A (MPLA) (18), a modified TLR4 agonist with less toxicity than LPS. ...
Article
Full-text available
With the advent of combined immunotherapies, personalized dendritic cell (DC)-based vaccination could integrate the current standard of care for the treatment of a large variety of tumors. Due to their proficiency at antigen presentation, DC are key coordinators of the innate and adaptive immune system, and have critical roles in the induction of antitumor immunity. However, despite proven immunogenicity and favorable safety profiles, DC-based immunotherapies have not succeeded at inducing significant objective clinical responses. Emerging data suggest that the combination of DC-based vaccination with other cancer therapies may fully unleash the potential of DC-based cancer vaccines and improve patient survival. In this review, we discuss the recent efforts to develop innovative personalized DC-based vaccines and their use in combined therapies, with a particular focus on ovarian cancer and the promising results of mutanome-based personalized immunotherapies.