PGE2, an essential homeostatic factor, is also a key mediator of immunopathology in chronic infections and cancer. The impact of PGE2 reflects the balance between its cyclooxygenase 2-regulated synthesis and 15-hydroxyprostaglandin dehydrogenase-driven degradation and the pattern of expression of PGE2 receptors. PGE2 enhances its own production but suppresses acute inflammatory mediators, resulting in its predominance at late/chronic stages of immunity. PGE2 supports activation of dendritic cells but suppresses their ability to attract naive, memory, and effector T cells. PGE2 selectively suppresses effector functions of macrophages and neutrophils and the Th1-, CTL-, and NK cell-mediated type 1 immunity, but it promotes Th2, Th17, and regulatory T cell responses. PGE2 modulates chemokine production, inhibiting the attraction of proinflammatory cells while enhancing local accumulation of regulatory T cells cells and myeloid-derived suppressor cells. Targeting the production, degradation, and responsiveness to PGE2 provides tools to modulate the patterns of immunity in a wide range of diseases, from autoimmunity to cancer.

Prostaglandins are small-molecule derivatives of arachidonic acid (AA), produced by cyclooxygenases (COX; constitutively active cyclooxygenase COX1 and inducible COX2) and PG synthases (1), with a relatively minor contribution of the isoprostane pathway (2). Local levels of PGE2, the main product of cyclooxygenases in myeloid and stromal cells, are regulated by the local balance between the COX2-driven synthesis and 15-hydroxyprostaglandin dehydrogenase (15-PGDH)-mediated degradation of PGE2 (1, 3). The receptors for PGE2 (EP1–EP4) are present on multiple cell types (4), reflecting the ubiquitous functions of PGE2, which span nociception and other aspects of neuronal signaling, hematopoiesis, regulation of blood flow, renal filtration and blood pressure, regulation of mucosal integrity, vascular permeability, and smooth muscle function (59). The present review focuses on the role of PGE2 and its receptors in the regulation of different stages of immune responses and different effector mechanisms of immunity.

PGE2 (molecular mass of 352 Da), recognized as a biologically active factor in the 1960s, has been shown to regulate multiple aspects of inflammation and multiple functions of different immune cells (1). Although generally recognized as a mediator of active inflammation, promoting local vasodilatation and local attraction and activation of neutrophils, macrophages, and mast cells at early stages of inflammation (1013), its ability to promote the induction of suppressive IL-10 and to directly suppress the production of multiple proinflammatory cytokines allow it to limit nonspecific inflammation, promoting the immune suppression associated with chronic inflammation and cancer (1, 14). Although PGE2 can promote the activation, maturation, and migration of dendritic cells (DCs) (see below), the central cells during the development of Ag-specific immunity, it has been widely demonstrated to suppress both innate and Ag-specific immunity at multiple molecular and cellular levels (1, 15), earning PGE2 the paradoxical status of a proinflammatory factor with immunosuppressive activity.

Although PGE2 inhibitors, such as steroids (inhibitors of AA release) and nonsteroid anti-inflammatory drugs (blockers of COX1/2 or COX2 function), represent some of the most common and effective pharmaceutical agents, realizing the full potential of PGE2 targeting in the treatment of chronic infections, inflammation, and cancer is restricted by the complex pattern of PGE2-mediated immunoregulation and our still incomplete understanding of the key mechanisms and targets of PGE2-mediated immunoregulation.

Regulation of PGE2 production

PGE2 can be produced by all cell types of the body, with epithelia, fibroblasts, and infiltrating inflammatory cells representing the major sources of PGE2 in the course of an immune response. The process of PGE2 synthesis involves phospholipase A2 family members that mobilize AA from cellular membranes (16), cyclooxygenases (constitutively active COX1 and inducible COX2) that convert AA into PGH2, and PGE synthases, needed for the final formulation of PGE2 (17) (Fig. 1). Although the rate of PGE2 synthesis and the resulting inflammatory process can be affected by additional factors, such as local availability of AA, in most physiologic conditions the rate of PGE2 synthesis is controlled by local expression and activity of COX2.

FIGURE 1.

Regulation of PGE2 synthesis, degradation, and responsiveness to PGE2. PGE2 synthesis is initiated by the (glucocorticosteroids sensitive) phospholipase A2-driven release of AA from cell membranes. AA becomes the substrate for COX1 (constitutive activity) and COX2 (inducible) that convert AA to PGH2 (process that can be suppressed by nonsteroid anti-inflammatory drugs), which is then converted to biologically active PGE2 by PGE synthases. PGE2 signals via four known receptors (EP1–EP4), with the cAMP/PKA/CREB signaling pathway responsible for major suppressive and regulatory functions of PGE2. Local PGE2 degradation is regulated by 15-PGDH. Dark-green arrows indicate currently applied inhibitory drugs; light green arrows indicate potential targets for prospective drugs. (+), activating; (−), inhibitory.

FIGURE 1.

Regulation of PGE2 synthesis, degradation, and responsiveness to PGE2. PGE2 synthesis is initiated by the (glucocorticosteroids sensitive) phospholipase A2-driven release of AA from cell membranes. AA becomes the substrate for COX1 (constitutive activity) and COX2 (inducible) that convert AA to PGH2 (process that can be suppressed by nonsteroid anti-inflammatory drugs), which is then converted to biologically active PGE2 by PGE synthases. PGE2 signals via four known receptors (EP1–EP4), with the cAMP/PKA/CREB signaling pathway responsible for major suppressive and regulatory functions of PGE2. Local PGE2 degradation is regulated by 15-PGDH. Dark-green arrows indicate currently applied inhibitory drugs; light green arrows indicate potential targets for prospective drugs. (+), activating; (−), inhibitory.

Close modal

Regulation of PGE2 degradation

PGE2 is relatively stable in vitro although its decay is accelerated by albumin (18). In contrast, PGE2 has a very rapid turnover rate in vivo and is rapidly eliminated from tissues and circulation (19). The rate of PGE2 degradation in individual tissues is controlled by 15-PGDH (3). The suppression of 15-PGDH activity is observed in many forms of cancer (2024) or UV-irradiated skin (25), the PGE2-rich and immunosuppressive environments. Apoptotic cancer cells can modulate the prostanoid produc-tion by enhancing the macrophage expression of COX2 and microsomal PGE synthase-1 while suppressing 15-PGDH (26). Moreover, the deactivation of 15-PGDH has been shown responsible for the resistance of premalignant colon lesions to celecoxib (24). These observations suggest that in addition to the rate of PGE2 synthesis, the rate of PGE2 decay may contribute to immune pathology and constitute a potential target for immunomodulation (21).

PGE2 receptors and signaling pathways: regulation of PGE2 responsiveness

The heterogeneous effects of PGE2 are reflected by the existence of four different PGE2 receptors, designated EP1, EP2, EP3 and EP4, with an additional level of functional diversity resulting from multiple splice variants of EP3 that exists in at least eight forms in humans and three forms in mice (reviewed in Ref. 4).

EP3 and EP4 represent high-affinity receptors, whereas EP1 and EP2 require significantly higher concentrations of PGE2 for effective signaling. The signaling through the two Gs-coupled receptors, EP2 and EP4, is mediated by the adenylate cyclase-triggered cAMP/PKA/CREB pathway (2729), mediating the dominant aspects of the anti-inflammatory and suppressive activity of PGE2 (Fig. 1). Despite their similar nominal functions, the signaling by EP2 and EP4 is triggered by different concentrations of PGE2 and differs in duration. EP4 signaling is rapidly desensitized following its PGE2 interaction, whereas EP2 is resistant to ligand-induced desensitization, implicating its ability to mediate PGE2 functions over prolonged periods of time and at later time points of inflammation (30). Although EP2 is thought to signal in a largely cAMP-dependent fashion, EP4 also activates the PI3K-dependent ERK1/2 pathway (31). However, both EP2 and EP4 have been shown to activate the GSK3/β-catenin pathway (32).

In contrast to EP2 and EP4, low-affinity EP1 and high-affinity EP3 are not coupled to Gs and lack cAMP-activating functions. Most of the splice variants of EP3 represent Gi-coupled PGE2 receptors that inhibit adenylate cyclase (33), although at least some are Gs-coupled and show different sensitivity to ligand-induced desensitization (4). Signaling via EP1 involves calcium release (4).

The differences in sensitivity, susceptibility to desensitization, and ability to activate different signaling pathways between the different PGE2 receptor systems allow for adaptable patterns of responses of different cell types at different stages of immune responses. Additional flexibility of the PGE2 receptor system results from different sensitivity of the individual receptors to regulation by additional factors. The expression of EP2 and the resulting responsiveness to PGE2 can be suppressed by hypermethylation, as observed in patients with idiopathic lung fibrosis (34). These observations raise the possibility that, in addition to the regulation of PGE2 production and its degradation, the regulation of PGE2 responsiveness at the level of expression of individual PGE2 receptors can also contribute to the pathogenesis of human disease and be exploited in their therapy. In support of this possibility, the use of synthetic inhibitors, preferentially affecting EP2, EP3, or EP4 signaling, allows for differential suppression of different aspects of PGE2 activity (reviewed in Ref. 4).

Although PGE2 can promote the tissue influx of neutrophils (10), macrophages (11), and mast cells (13), it differentially affects the functions of different innate effector cells.

NK cells

PGE2 suppresses the cytolytic effector functions of NK cells (35, 36), in a mechanism involving suppression of IL-12 and IL-15 responsiveness (37, 38), and most likely IL-2. It also inhibits NK cell production of IFN-γ, abrogating NK cell “helper” function in the DC-mediated induction of Th1 and CTL responses (39). PGE2-mediated suppression of NK cell function during surgery has been shown to facilitate the establishment of metastases in experimental animals (40).

Granulocytes

PGE2 has been shown to inhibit granulocyte functions (41), contributing to the defective innate host defense in patients after bone marrow transplantation or with cancer, as well as other conditions associated with overproduction of PGE2 (42).

Macrophages

Acting in an EP2-dependent (43) and PTEN-dependent (44) manner, PGE2 limits the phagocytosis by alveolar macrophages (43) and their pathogen-killing function (45). At least a part of the inhibitory impact of PGE2 on the alveolar macrophage function is mediated via the induction of IL-1R–associated kinase-M, which blocks the scavenger receptor-mediated phagocytosis and the TLR-dependent activation of TNF-α (46).

Mast cells

PGE2 promotes both the induction of mast cells (47) and their local attraction and degranulation in a mechanism involving EP1 and EP3 (1113, 48). It also promotes the degranulation-independent production of the proangiogenic and immunosuppressive vascular endothelial growth factor and MCP-1 by mast cells (11, 49), which contributes to the overall disease-promoting activity of PGE2 in cancer.

PGE2 affects several key phenomena relevant to the induction of immune responses. In addition to its multifaceted regulation of DC functions during the priming of naive T cells (see below), it also directly inhibits T cell production of IL-2 (50) and IL-2 responsiveness (51), suppressing the activation and expansion of Ag-specific T cells.

DC development

PGE2 has been shown to disrupt early stages of DC differentiation (52), contributing to local and systemic DC dysfunction in cancer (5355) or following UV exposure (56, 57). Although the ability of PGE2 to suppress the differentiation of functionally competent Th1-inducing DCs has been long recognized (52), it was recently shown that the resulting “PGE2 DCs” represent myeloid-derived suppressor cells (MDSCs), capable of suppressing CTL responses (58).

DC activation, migration, and stimulatory function

In striking contrast to its uniform inhibitory impact on early stages of DC development, PGE2 has a much more complex effect on the activation of fully developed (although functionally immature) DCs. PGE2 has been shown to support the induction of fully mature DCs capable of homing to lymph nodes and to be highly effective in priming naive T cells. The addition of PGE2 to the mixture of proinflammatory cytokines involving IL-1β and TNF-α accelerates DC maturation and elevates their expression of costimulatory molecules (5961).

PGE2 has been shown to promote high-level expression of CCR7 (the receptor for CCL19 and CCL21) and responsiveness to these lymph node-type chemokines in maturing monocyte-derived DCs (62, 63). This activity and its roles in podosome dissolution (64) and induction of matrix metalloproteinase-9 (65) suggested the role for PGE2 in DC migration to the lymph nodes. However, recent data demonstrate that the CCR7-enhancing effects of PGE2 are mediated by the suppression of CCL19 in maturing DCs (endogenous CCR7 ligand driving CCR7 internalization) and are rapidly compensated after DC removal from the maturation cultures (66). An additional factor that can limit the in vivo migratory potential of the PGE2-matured DCs is the ability of PGE2 to induce tissue inhibitor of proteinase-1 (67).

Although PGE2-matured DCs indeed migrate to the lymph nodes faster than do immature DCs (68), two small clinical studies comparing the in vivo migratory capacity of differentially matured human DCs did not reveal any migratory advantage of DCs conferred by exogenous PGE2 (66, 69). In line with the notion that DC maturation and effective lymph node migration can occur in the absence of PGE2, a recent mouse study in PGE synthase-1–deficient animals showed an abrogation of PGE2 synthesis by DCs and their altered cytokine profile, but it did not reveal any impact on their maturation status or migratory function (70). These data suggest that although PGE2 can enhance the migratory function of DCs, it is not critically required in this regard and can be successfully replaced by alternative factors.

The elevated expression of the maturation-associated costimulatory factors on the surface of PGE2-matured DCs translates into their enhanced ability to activate naive T cells, when compared with immature DCs (5961). Although PGE2 also enhances the DC production of several suppressive factors, such as IL-10 (52), thrombospondin-1, (71), and IDO (72), its most frequently observed net effect during DC maturation is the enhanced ability of DCs to promote T cell expansion (5961).

However, DCs matured in the presence of PGE2 develop a distinct “exhausted” phenotype, manifested by their impaired ability (compared with alternatively matured DCs) to induce the CTL-, Th1-, and NK cell-mediated type 1 immunity (61, 73, 74), while promoting Th2 responses (73). Such negative effects are mediated by the suppression of proinflammatory cytokines, including the bioactive IL-12p70 (61) (see below). In accordance with the notion that exogenous PGE2 may have a net inhibitory effect on the functional activity of maturing DCs, it was shown that the replacement of PGE2 by other DC maturation-driving factors can enhance the immunogenic and anti-tumor effectiveness of DC vaccines (75, 76).

Regulation of the attraction of naive T cells, DC–T cell interaction, and T cell activation

CCR7 ligands (CCL19/MIP3β/ELC and CCL21/6Ckine/SLC) and CXCR4 ligand CXCL12/SDF-1 represent two groups of chemokines needed for effective T cell entry into lymph nodes (77). Although the role of PGE2 in the local regulation of these two chemokines within the lymph nodes remains unclear, PGE2 has been recently shown to suppress the ability of DCs to produce CCL19 (the only CCR7 ligand produced by human monocyte-derived DCs) and to block the ability of DCs to attract naive T cells (66). In contrast, PGE2 was shown to enhance the production of CXCL12 by vascular endothelium (78), raising the possibility that a similar effect may also operate in the lymph nodes, resulting in an opposite impact of PGE2 on the CCR7- versus CXCR4-driven events governing T cell accumulation in the lymph nodes and their interaction with different types of APCs.

The suppressive effects of PGE2 on the activation and expansion of naive T cells also include the direct inhibitory effects of PGE2 on IL-2 production (50) and the expression of IL-2 receptor and JAK3, which mediate the responsiveness of T cells to IL-2 (51, 79).

In accord with the overall suppressive, rather than stimulatory, impact of PGE2 during the induction of immune responses, the suppression of COX2 activity during vaccination was shown to enhance the immune and therapeutic activity of cancer vaccines (80, 81).

PGE2 suppress IL-2 production and IL-2 responsiveness in T cells, nonspecifically suppressing T cell activation and proliferation at high doses. Already much lower concentrations of PGE2 show profound modulatory effects shifting the pattern of CD4+ T cell responses from the aggressive Th1 cells (promoting the inflammatory/cytotoxic form of immunity) toward Th2 and Th17 cells that mediate less tissue-destructive forms of immunity.

Balance between Th1 and Th2 responses

The original evidence that PGE2 is involved in regulating the balance between different forms of Th cell responses came from in vitro studies showing its ability to selectively inhibit the production of the Th1 cytokine IFN-γ, but not the Th2 cytokines IL-4 and IL-5, in mouse (82) and human CD4+ T cells (83).

In addition to its direct impact on CD4+ T cells, the Th1-suppressive impact of PGE2 also relies on its ability to suppress the production of IL-12 (84) in monocytes (84) and DCs (52, 61). Additional mechanisms of the IL-12 antagonistic activity of PGE2 include its ability to suppress the expression of IL-12 receptor and the resulting responsiveness to IL-12 (85), and may include the induction of IL-12p40 homodimer (86, 87), a competitive inhibitor of the IL-12 receptor in mice (88). Thus, PGE2 shifts the balance away from Th1 responses toward other forms of immunity, such as Th2 responses. In support of its involvement in Th2-mediated human pathology, overproduction of PGE2 is observed in multiple Th2-associated diseases, most notably atopic dermatitis and asthma (89).

Th17 differentiation

EP2- and EP4-dependent signals from PGE2 have also been shown to promote the development of IL-17–producing T cells in multiple models of infection and autoimmunity (9093). The Th17-promoting activity of PGE2 is related to its ability to suppress the production of (Th17-inhibitory) IL-12p70 while enhancing the Th17-supporting IL-23 (94).

CTL differentiation and effector function

Mouse models demonstrated that the induction of CTL activity against viral Ags and alloantigens is highly sensitive to PGE2 and cAMP elevation (95, 96). PGE2-dependent inhibition of CTL activity contributes to local immune suppression in decidual tissues and tumors (9799). Interestingly, apart from the interference with the de novo development of CTL activity, PGE2 can also suppress the ability of fully developed CTLs to interact with their targets and kill tumor cells (99, 100). In addition to its direct effects on CD8+ T cells, PGE2 has also been shown to suppress the ability of maturing DCs to develop CTL-inducing function by suppressing their ability to secrete IL-12 during the subsequent interaction with naive CD8+ T cells (101).

Interestingly, CTLs can produce PGE2 by themselves, resulting in the acquisition of their suppressive function (102), although the implications of this phenomenon to the overall regulation of CTL cell function remain unclear.

B cells

PGE2 has been shown to interfere with early stages of B cell activation and show profound cAMP-mediated regulation of the process of Ig class switch in activated B cells (1, 103). Perhaps the most striking of these effects is the ability of PGE2 to promote IgE production, the phenomenon contributing to atopic diseases (104), jointly with the ability of PGE2 to support the induction, attraction, and degranulation of mast cells (1113, 47, 48).

In addition to its long-recognized direct inhibitory effects on type 1 immune cells, more recent studies demonstrate indirect suppressive effects of PGE2, enhancing the development and activity of suppressive types of immune cells.

Regulatory T cell activity

PGE2 has been shown to promote the development of regulatory T cells (Tregs) in humans and in mice (105108). COX2 and PGE2 have been shown to be essential for the EP2- and EP4-dependent induction of murine Tregs in cancer (106) and following skin UV irradiation (108), with an analogous role demonstrated in human tumor tissues (107). In addition to promoting de novo Treg differentiation from naive precursors, PGE2 also promotes the interaction of DCs with Tregs (109), suggesting that it may also promote the expansion of pre-existing Tregs, as observed in cancer patients vaccinated with PGE2-matured DCs (110). Interestingly, PGE2 is also involved in mediating the suppressive activity of Tregs (111).

Suppressive macrophages and myeloid-derived suppressor cells: positive and negative feedback involving PGE2

PGE2 is needed for the development of tumor-associated suppressive macrophages (55, 112, 113) and myeloid-derived suppressor cells (58, 114116). Interestingly, in addition to being the recipients of PGE2-mediated signals, MDSCs express high levels of COX2 and are a major source of PGE2 secretion in human cancer (58, 117). The resulting positive feedback loop between PGE2 and COX2 is essential for the functional stability of MDSCs, as well as their ability to produce the additional MDSC-associated suppressive mediators and to suppress CD8+ T cell function (58). Because PGE2 participates in the induction of hypoxia-inducible factor-1α (118), the hypoxia-inducible factor-1α–mediated development of MDSCs (119) is likely to represent a central downstream signaling event in the PGE2-mediated impact on MDSC development.

PGE2 has been also shown to be critical for the development of the apoptotic body-induced suppressor function in macrophages, promoting the growth of intracellular parasites (120). Interestingly, PGE2 is a known inducer of another suppressive factor, IL-10, in tissue macrophages (112, 113). IL-10 acts as a controller of PGE2 secretion, resulting in the paradoxical role of IL-10 in the reversal of the PGE2-mediated macrophage dysfunction, facilitating effective control of the infection with pathogenic strain of Escherichia coli (121).

In addition to its opposite impact on the development and function of the effector versus suppressive cells, PGE2 also differentially regulates their influx to affected tissues. PGE2 enhances the production of CXCL8/IL-8, the attractant for neutrophils (10) and macrophage-recruiting CCL2/MCP-1 (11). It is also a chemoattractant for mast cells (13), helping to recruit the three members of innate immune system specialized in fighting extracellular pathogens at early stages of immune responses.

However, the macrophage-attracting properties of PGE2 are limited by its ability to block the expression of CCR5 and Mac-1 on monocytes and macrophages, leading to interference with their extravasation and functions (122). The PGE2-driven suppression of CCL5, as well as all three CXCR3 ligands, CXCL9/MIG, CXCL10/ IP10, and CXCL11/ITAC, results in its powerful inhibition of the attraction of not only the proinflammatory-type macrophages but also the CCR5+ and CXCR3+ type 1 effector cells (CTLs, NK cells, and Th1 cells) (74, 109, 123, 124). At the same time, PGE2 enhances the production of Th2-attracting chemokines (124) and promotes the production of CCL22/MDC and the resulting attraction of Tregs (109), as well as the CXCL12/SDF1-driven accumulation of MDSCs (125).

In addition to the differential regulation of the effector and Treg/MDSC-attracting chemokines, PGE2 also interferes with the expression of chemokine receptors. It blocks the induction of CCR5 on monocytes (122) and suppresses the DC and IL-12–driven induction of CCR5 and CXCR3 on CD8+ T cells (101), whereas it induces and stabilizes the expression of CXCR4 on cancer-associated MDSCs (125). Additionally, PGE2 is known to suppress the ability of gut-associated DCs to produce retinoic acid, needed for the ability of DCs to induce the CCR9 expression and gut-homing function in responding T cells (126).

PGE2 has also been shown to block the transendothelial migration of human and murine T lymphocytes, interfering with the expression and functions of relevant integrins (127, 128) and directly suppressing CTL motility (100).

In brief, PGE2 supports acute local inflammation and phagocyte-mediated immunity at the site of pathogen entry, but it has a specialized role in controlling the potentially harmful activation of CTL-, Th1-, and NK cell-mediated type 1 (cytotoxic) immunity, especially at later stages of immune responses (Fig. 2). Such PGE2-mediated suppression of type 1 immunity by PGE2 shifts the pattern of immune reactivity toward a less aggressive form of immunity mediated by Th2 and Th17 cells as well as B cells, as well as enhancement of the Treg- and MDSC-mediated suppressive events.

FIGURE 2.

Regulation of the immune responses by PGE2. PGE2 supports local acute inflammation and phagocyte-mediated immunity at the site of pathogen entry while selectively suppressing the CTL-, Th1-, and NK cell-mediated type 1 (cytotoxic) forms of immunity at the stage of their induction in lymphoid tissues and by differentially regulating the influx and activity of the effector versus regulatory cells into affected tissues. Blue indicates relevant to immunity against intracellular pathogens and cancer; green indicates relevant to immunity against extracellular pathogens; purple indicates relevant to immune suppression. ↓, suppression; ↑, enhancement; MC, mast cells; Mf, macrophages; N, neutrophils.

FIGURE 2.

Regulation of the immune responses by PGE2. PGE2 supports local acute inflammation and phagocyte-mediated immunity at the site of pathogen entry while selectively suppressing the CTL-, Th1-, and NK cell-mediated type 1 (cytotoxic) forms of immunity at the stage of their induction in lymphoid tissues and by differentially regulating the influx and activity of the effector versus regulatory cells into affected tissues. Blue indicates relevant to immunity against intracellular pathogens and cancer; green indicates relevant to immunity against extracellular pathogens; purple indicates relevant to immune suppression. ↓, suppression; ↑, enhancement; MC, mast cells; Mf, macrophages; N, neutrophils.

Close modal

Although PGE2 can accelerate DC maturation and migratory function, the PGE2-dependent suppression of the naive T cell-attracting CCL19 in DCs (66) and its suppression of IL-2 and IL-12 production and functions result in the net inhibitory activity of PGE2 during the induction of Ag-specific immunity, reflected by the ability of COX2 inhibitors to enhance the immune and therapeutic activity of cancer vaccines (80, 81). In accordance with its selective functions in regulating the effector phase of immunity, PGE2 potently suppresses type 1 effector mechanisms, playing an important role of tissue preservation in such critical organs as the lung (34, 129) and reproductive system (6), while shifting the pattern of T cell responses toward Th2, Th17, and Treg activity, helping to contain the damage to tissues during prolonged immune responses.

Although this ability of PGE2 to limit type 1 immunity is crucial for the host self-preservation, it is counterproductive during infections with intracellular organisms (such as mycobacteria or HIV) and in cancer, which both depend on enhanced PGE2 production and/or reduced degradation of PGE2 for the establishment of immunosuppression and disease progression. Although the therapeutic antagonism with the PGE2 system has traditionally focused on the inhibition of PGE2 production using nonselective or COX2-selective blockers, the newly available agonists and antagonists of the individual PGE2 receptors, as well as the new understanding of the key role of 15-PGDH in controlling PGE2 degradation in the tissues, allow for new therapeutic approaches to control the PGE2-mediated immunopathology. Additionally, amplification of PGE2 production and responsiveness to this factor and antagonizing its rate of decay may be used to treat autoimmune phenomena. Recent advances and prospective identification of key mechanisms regulating the functions of 15-PGDH and individual PGE2 receptors in different organs and cell types (and the balance between their different signaling pathways) are likely to result in new therapeutic strategies with higher potency and improved selectivity of action.

I thank Drs. Natasa Obermajer, Ravikumar Muthuswamy, Erik Berk, and Jeffrey Wong for stimulating discussions and comments.

This work was supported by National Institutes of Health Grants 1PO1CA132714 and 1P50CA121973.

Abbreviations used in this article:

AA

arachidonic acid

COX

cyclooxygenase

DC

dendritic cell

MDSC

myeloid-derived suppressor cell

15-PGDH

15-hydroxyprostaglandin dehydrogenase

Treg

regulatory T cell.

1
Phipps
R. P.
,
Stein
S. H.
,
Roper
R. L.
.
1991
.
A new view of prostaglandin E regulation of the immune response.
Immunol. Today
12
:
349
352
.
2
Gao
L.
,
Zackert
W. E.
,
Hasford
J. J.
,
Danekis
M. E.
,
Milne
G. L.
,
Remmert
C.
,
Reese
J.
,
Yin
H.
,
Tai
H. H.
,
Dey
S. K.
, et al
.
2003
.
Formation of prostaglandins E2 and D2 via the isoprostane pathway: a mechanism for the generation of bioactive prostaglandins independent of cyclooxygenase.
J. Biol. Chem.
278
:
28479
28489
.
3
Tai
H. H.
,
Ensor
C. M.
,
Tong
M.
,
Zhou
H.
,
Yan
F.
.
2002
.
Prostaglandin catabolizing enzymes.
Prostaglandins Other Lipid Mediat.
68-69
:
483
493
.
4
Hata
A. N.
,
Breyer
R. M.
.
2004
.
Pharmacology and signaling of prostaglandin receptors: multiple roles in inflammation and immune modulation.
Pharmacol. Ther.
103
:
147
166
.
5
Durand
E. M.
,
Zon
L. I.
.
2010
.
Newly emerging roles for prostaglandin E2 regulation of hematopoiesis and hematopoietic stem cell engraftment.
Curr. Opin. Hematol.
17
:
308
312
.
6
Nagamatsu
T.
,
Schust
D. J.
.
2010
.
The immunomodulatory roles of macrophages at the maternal-fetal interface.
Reprod. Sci.
17
:
209
218
.
7
Rivest
S.
2010
.
Interactions between the immune and neuroendocrine systems.
Prog. Brain Res.
181
:
43
53
.
8
Yuhki
K.
,
Kashiwagi
H.
,
Kojima
F.
,
Kawabe
J.
,
Ushikubi
F.
.
2010
.
Roles of prostanoids in the pathogenesis of cardiovascular diseases.
Int. Angiol.
29
(
Suppl.
):
19
27
.
9
Yang
H.
,
Chen
C.
.
2008
.
Cyclooxygenase-2 in synaptic signaling.
Curr. Pharm. Des.
14
:
1443
1451
.
10
Yu
Y.
,
Chadee
K.
.
1998
.
Prostaglandin E2 stimulates IL-8 gene expression in human colonic epithelial cells by a posttranscriptional mechanism.
J. Immunol.
161
:
3746
3752
.
11
Nakayama
T.
,
Mutsuga
N.
,
Yao
L.
,
Tosato
G.
.
2006
.
Prostaglandin E2 promotes degranulation-independent release of MCP-1 from mast cells.
J. Leukoc. Biol.
79
:
95
104
.
12
Wang
X. S.
,
Lau
H. Y.
.
2006
.
Prostaglandin E potentiates the immunologically stimulated histamine release from human peripheral blood-derived mast cells through EP1/EP3 receptors.
Allergy
61
:
503
506
.
13
Weller
C. L.
,
Collington
S. J.
,
Hartnell
A.
,
Conroy
D. M.
,
Kaise
T.
,
Barker
J. E.
,
Wilson
M. S.
,
Taylor
G. W.
,
Jose
P. J.
,
Williams
T. J.
.
2007
.
Chemotactic action of prostaglandin E2 on mouse mast cells acting via the PGE2 receptor 3.
Proc. Natl. Acad. Sci. USA
104
:
11712
11717
.
14
Wang
M. T.
,
Honn
K. V.
,
Nie
D.
.
2007
.
Cyclooxygenases, prostanoids, and tumor progression.
Cancer Metastasis Rev.
26
:
525
534
.
15
Harris
S. G.
,
Padilla
J.
,
Koumas
L.
,
Ray
D.
,
Phipps
R. P.
.
2002
.
Prostaglandins as modulators of immunity.
Trends Immunol.
23
:
144
150
.
16
Lambeau
G.
,
Lazdunski
M.
.
1999
.
Receptors for a growing family of secreted phospholipases A2.
Trends Pharmacol. Sci.
20
:
162
170
.
17
Park
J. Y.
,
Pillinger
M. H.
,
Abramson
S. B.
.
2006
.
Prostaglandin E2 synthesis and secretion: the role of PGE2 synthases.
Clin. Immunol.
119
:
229
240
.
18
Fitzpatrick
F. A.
,
Aguirre
R.
,
Pike
J. E.
,
Lincoln
F. H.
.
1980
.
The stability of 13,14-dihydro-15 keto-PGE2.
Prostaglandins
19
:
917
931
.
19
Förstermann
U.
,
Neufang
B.
.
1983
.
Elimination from the circulation of cats of 6-keto-prostaglandin E1 compared with prostaglandins E2 and I2.
J. Pharm. Pharmacol.
35
:
724
728
.
20
Backlund
M. G.
,
Mann
J. R.
,
Holla
V. R.
,
Buchanan
F. G.
,
Tai
H. H.
,
Musiek
E. S.
,
Milne
G. L.
,
Katkuri
S.
,
DuBois
R. N.
.
2005
.
15-Hydroxyprostaglandin dehydrogenase is down-regulated in colorectal cancer.
J. Biol. Chem.
280
:
3217
3223
.
21
Hirsch
F. R.
,
Lippman
S. M.
.
2005
.
Advances in the biology of lung cancer chemoprevention.
J. Clin. Oncol.
23
:
3186
3197
.
22
Tseng-Rogenski
S.
,
Gee
J.
,
Ignatoski
K. W.
,
Kunju
L. P.
,
Bucheit
A.
,
Kintner
H. J.
,
Morris
D.
,
Tallman
C.
,
Evron
J.
,
Wood
C. G.
, et al
.
2010
.
Loss of 15-hydroxyprostaglandin dehydrogenase expression contributes to bladder cancer progression.
Am. J. Pathol.
176
:
1462
1468
.
23
Eruslanov
E.
,
Kaliberov
S.
,
Daurkin
I.
,
Kaliberova
L.
,
Buchsbaum
D.
,
Vieweg
J.
,
Kusmartsev
S.
.
2009
.
Altered expression of 15-hydroxyprostaglandin dehydrogenase in tumor-infiltrated CD11b myeloid cells: a mechanism for immune evasion in cancer.
J. Immunol.
182
:
7548
7557
.
24
Yan
M.
,
Myung
S. J.
,
Fink
S. P.
,
Lawrence
E.
,
Lutterbaugh
J.
,
Yang
P.
,
Zhou
X.
,
Liu
D.
,
Rerko
R. M.
,
Willis
J.
, et al
.
2009
.
15-Hydroxyprostaglandin dehydrogenase inactivation as a mechanism of resistance to celecoxib chemoprevention of colon tumors.
Proc. Natl. Acad. Sci. USA
106
:
9409
9413
.
25
Judson
B. L.
,
Miyaki
A.
,
Kekatpure
V. D.
,
Du
B.
,
Gilleaudeau
P.
,
Sullivan-Whalen
M.
,
Mohebati
A.
,
Nair
S.
,
Boyle
J. O.
,
Granstein
R. D.
, et al
.
2010
.
UV radiation inhibits 15-hydroxyprostaglandin dehydrogenase levels in human skin: evidence of transcriptional suppression.
Cancer Prev. Res. (Phila.)
3
:
1104
1111
.
26
Brecht
K.
,
Weigert
A.
,
Hu
J.
,
Popp
R.
,
Fisslthaler
B.
,
Korff
T.
,
Fleming
I.
,
Geisslinger
G.
,
Brüne
B.
.
2011
.
Macrophages programmed by apoptotic cells promote angiogenesis via prostaglandin E2.
FASEB J.
25
:
2408
2417
.
27
Honda
A.
,
Sugimoto
Y.
,
Namba
T.
,
Watabe
A.
,
Irie
A.
,
Negishi
M.
,
Narumiya
S.
,
Ichikawa
A.
.
1993
.
Cloning and expression of a cDNA for mouse prostaglandin E receptor EP2 subtype.
J. Biol. Chem.
268
:
7759
7762
.
28
Regan
J. W.
,
Bailey
T. J.
,
Pepperl
D. J.
,
Pierce
K. L.
,
Bogardus
A. M.
,
Donello
J. E.
,
Fairbairn
C. E.
,
Kedzie
K. M.
,
Woodward
D. F.
,
Gil
D. W.
.
1994
.
Cloning of a novel human prostaglandin receptor with characteristics of the pharmacologically defined EP2 subtype.
Mol. Pharmacol.
46
:
213
220
.
29
Fujino
H.
,
Salvi
S.
,
Regan
J. W.
.
2005
.
Differential regulation of phosphorylation of the cAMP response element-binding protein after activation of EP2 and EP4 prostanoid receptors by prostaglandin E2.
Mol. Pharmacol.
68
:
251
259
.
30
Nishigaki
N.
,
Negishi
M.
,
Ichikawa
A.
.
1996
.
Two Gs-coupled prostaglandin E receptor subtypes, EP2 and EP4, differ in desensitization and sensitivity to the metabolic inactivation of the agonist.
Mol. Pharmacol.
50
:
1031
1037
.
31
Fujino
H.
,
Xu
W.
,
Regan
J. W.
.
2003
.
Prostaglandin E2 induced functional expression of early growth response factor-1 by EP4, but not EP2, prostanoid receptors via the phosphatidylinositol 3-kinase and extracellular signal-regulated kinases.
J. Biol. Chem.
278
:
12151
12156
.
32
Fujino
H.
,
West
K. A.
,
Regan
J. W.
.
2002
.
Phosphorylation of glycogen synthase kinase-3 and stimulation of T-cell factor signaling following activation of EP2 and EP4 prostanoid receptors by prostaglandin E2.
J. Biol. Chem.
277
:
2614
2619
.
33
Sugimoto
Y.
,
Namba
T.
,
Honda
A.
,
Hayashi
Y.
,
Negishi
M.
,
Ichikawa
A.
,
Narumiya
S.
.
1992
.
Cloning and expression of a cDNA for mouse prostaglandin E receptor EP3 subtype.
J. Biol. Chem.
267
:
6463
6466
.
34
Huang
S. K.
,
Fisher
A. S.
,
Scruggs
A. M.
,
White
E. S.
,
Hogaboam
C. M.
,
Richardson
B. C.
,
Peters-Golden
M.
.
2010
.
Hypermethylation of PTGER2 confers prostaglandin E2 resistance in fibrotic fibroblasts from humans and mice.
Am. J. Pathol.
177
:
2245
2255
.
35
Goto
T.
,
Herberman
R. B.
,
Maluish
A.
,
Strong
D. M.
.
1983
.
Cyclic AMP as a mediator of prostaglandin E-induced suppression of human natural killer cell activity.
J. Immunol.
130
:
1350
1355
.
36
Bankhurst
A. D.
1982
.
The modulation of human natural killer cell activity by prostaglandins.
J. Clin. Lab. Immunol.
7
:
85
91
.
37
Walker
W.
,
Rotondo
D.
.
2004
.
Prostaglandin E2 is a potent regulator of interleukin-12- and interleukin-18-induced natural killer cell interferon-γ synthesis.
Immunology
111
:
298
305
.
38
Joshi
P. C.
,
Zhou
X.
,
Cuchens
M.
,
Jones
Q.
.
2001
.
Prostaglandin E2 suppressed IL-15-mediated human NK cell function through down-regulation of common γ-chain.
J. Immunol.
166
:
885
891
.
39
Mailliard
R. B.
,
Alber
S. M.
,
Shen
H.
,
Watkins
S. C.
,
Kirkwood
J. M.
,
Herberman
R. B.
,
Kalinski
P.
.
2005
.
IL-18-induced CD83+CCR7+ NK helper cells.
J. Exp. Med.
202
:
941
953
.
40
Yakar
I.
,
Melamed
R.
,
Shakhar
G.
,
Shakhar
K.
,
Rosenne
E.
,
Abudarham
N.
,
Page
G. G.
,
Ben-Eliyahu
S.
.
2003
.
Prostaglandin E2 suppresses NK activity in vivo and promotes postoperative tumor metastasis in rats.
Ann. Surg. Oncol.
10
:
469
479
.
41
Smith
R. J.
1977
.
Modulation of phagocytosis by and lysosomal enzyme secretion from guinea-pig neutrophils: effect of nonsteroid anti-inflammatory agents and prostaglindins.
J. Pharmacol. Exp. Ther.
200
:
647
657
.
42
Ballinger
M. N.
,
Aronoff
D. M.
,
McMillan
T. R.
,
Cooke
K. R.
,
Olkiewicz
K.
,
Toews
G. B.
,
Peters-Golden
M.
,
Moore
B. B.
.
2006
.
Critical role of prostaglandin E2 overproduction in impaired pulmonary host response following bone marrow transplantation.
J. Immunol.
177
:
5499
5508
.
43
Aronoff
D. M.
,
Canetti
C.
,
Peters-Golden
M.
.
2004
.
Prostaglandin E2 inhibits alveolar macrophage phagocytosis through an E-prostanoid 2 receptor-mediated increase in intracellular cyclic AMP.
J. Immunol.
173
:
559
565
.
44
Canetti
C.
,
Serezani
C. H.
,
Atrasz
R. G.
,
White
E. S.
,
Aronoff
D. M.
,
Peters-Golden
M.
.
2007
.
Activation of phosphatase and tensin homolog on chromosome 10 mediates the inhibition of FcγR phagocytosis by prostaglandin E2 in alveolar macrophages.
J. Immunol.
179
:
8350
8356
.
45
Serezani
C. H.
,
Chung
J.
,
Ballinger
M. N.
,
Moore
B. B.
,
Aronoff
D. M.
,
Peters-Golden
M.
.
2007
.
Prostaglandin E2 suppresses bacterial killing in alveolar macrophages by inhibiting NADPH oxidase.
Am. J. Respir. Cell Mol. Biol.
37
:
562
570
.
46
Hubbard
L. L.
,
Ballinger
M. N.
,
Thomas
P. E.
,
Wilke
C. A.
,
Standiford
T. J.
,
Kobayashi
K. S.
,
Flavell
R. A.
,
Moore
B. B.
.
2010
.
A role for IL-1 receptor-associated kinase-M in prostaglandin E2-induced immunosuppression post-bone marrow transplantation.
J. Immunol.
184
:
6299
6308
.
47
Hu
Z. Q.
,
Asano
K.
,
Seki
H.
,
Shimamura
T.
.
1995
.
An essential role of prostaglandin E on mouse mast cell induction.
J. Immunol.
155
:
2134
2142
.
48
Gomi
K.
,
Zhu
F. G.
,
Marshall
J. S.
.
2000
.
Prostaglandin E2 selectively enhances the IgE-mediated production of IL-6 and granulocyte-macrophage colony-stimulating factor by mast cells through an EP1/EP3-dependent mechanism.
J. Immunol.
165
:
6545
6552
.
49
Abdel-Majid
R. M.
,
Marshall
J. S.
.
2004
.
Prostaglandin E2 induces degranulation-independent production of vascular endothelial growth factor by human mast cells.
J. Immunol.
172
:
1227
1236
.
50
Walker
C.
,
Kristensen
F.
,
Bettens
F.
,
deWeck
A. L.
.
1983
.
Lymphokine regulation of activated (G1) lymphocytes, I: Prostaglandin E2-induced inhibition of interleukin 2 production.
J. Immunol.
130
:
1770
1773
.
51
Rincón
M.
,
Tugores
A.
,
López-Rivas
A.
,
Silva
A.
,
Alonso
M.
,
De Landázuri
M. O.
,
López-Botet
M.
.
1988
.
Prostaglandin E2 and the increase of intracellular cAMP inhibit the expression of interleukin 2 receptors in human T cells.
Eur. J. Immunol.
18
:
1791
1796
.
52
Kaliński
P.
,
Hilkens
C. M.
,
Snijders
A.
,
Snijdewint
F. G.
,
Kapsenberg
M. L.
.
1997
.
IL-12-deficient dendritic cells, generated in the presence of prostaglandin E2, promote type 2 cytokine production in maturing human naive T helper cells.
J. Immunol.
159
:
28
35
.
53
Sharma
S.
,
Stolina
M.
,
Yang
S. C.
,
Baratelli
F.
,
Lin
J. F.
,
Atianzar
K.
,
Luo
J.
,
Zhu
L.
,
Lin
Y.
,
Huang
M.
, et al
.
2003
.
Tumor cyclooxygenase 2-dependent suppression of dendritic cell function.
Clin. Cancer Res.
9
:
961
968
.
54
Sombroek
C. C.
,
Stam
A. G.
,
Masterson
A. J.
,
Lougheed
S. M.
,
Schakel
M. J.
,
Meijer
C. J.
,
Pinedo
H. M.
,
van den Eertwegh
A. J.
,
Scheper
R. J.
,
de Gruijl
T. D.
.
2002
.
Prostanoids play a major role in the primary tumor-induced inhibition of dendritic cell differentiation.
J. Immunol.
168
:
4333
4343
.
55
Heusinkveld
M.
,
de Vos van Steenwijk
P. J.
,
Goedemans
R.
,
Ramwadhdoebe
T. H.
,
Gorter
A.
,
Welters
M. J.
,
van Hall
T.
,
van der Burg
S. H.
.
2011
.
M2 macrophages induced by prostaglandin E2 and IL-6 from cervical carcinoma are switched to activated M1 macrophages by CD4+ Th1 cells.
J. Immunol.
187
:
1157
1165
.
56
Lim
H. W.
,
Thorbecke
G. J.
,
Baer
R. L.
,
Gigli
I.
.
1983
.
Effect of indomethacin on alteration of ATPase-positive Langerhans cell density and cutaneous sunburn reaction induced by ultraviolet-B radiation.
J. Invest. Dermatol.
81
:
455
458
.
57
Ng
R. L.
,
Bisley
J. L.
,
Gorman
S.
,
Norval
M.
,
Hart
P. H.
.
2010
.
Ultraviolet irradiation of mice reduces the competency of bone marrow-derived CD11c+ cells via an indomethacin-inhibitable pathway.
J. Immunol.
185
:
7207
7215
.
58
Obermajer
N.
,
Muthuswamy
R.
,
Lesnock
J.
,
Edwards
R. P.
,
Kalinski
P.
.
2011
.
Positive feedback between PGE2 and COX2 redirects the differentiation of human dendritic cells towards stable myeloid-derived suppressor cells.
Blood
.
118
:
5498
5505
.
59
Jonuleit
H.
,
Kühn
U.
,
Müller
G.
,
Steinbrink
K.
,
Paragnik
L.
,
Schmitt
E.
,
Knop
J.
,
Enk
A. H.
.
1997
.
Pro-inflammatory cytokines and prostaglandins induce maturation of potent immunostimulatory dendritic cells under fetal calf serum-free conditions.
Eur. J. Immunol.
27
:
3135
3142
.
60
Rieser
C.
,
Böck
G.
,
Klocker
H.
,
Bartsch
G.
,
Thurnher
M.
.
1997
.
Prostaglandin E2 and tumor necrosis factor α cooperate to activate human dendritic cells: synergistic activation of interleukin 12 production.
J. Exp. Med.
186
:
1603
1608
.
61
Kaliński
P.
,
Schuitemaker
J. H.
,
Hilkens
C. M.
,
Kapsenberg
M. L.
.
1998
.
Prostaglandin E2 induces the final maturation of IL-12-deficient CD1a+CD83+ dendritic cells: the levels of IL-12 are determined during the final dendritic cell maturation and are resistant to further modulation.
J. Immunol.
161
:
2804
2809
.
62
Luft
T.
,
Jefford
M.
,
Luetjens
P.
,
Toy
T.
,
Hochrein
H.
,
Masterman
K. A.
,
Maliszewski
C.
,
Shortman
K.
,
Cebon
J.
,
Maraskovsky
E.
.
2002
.
Functionally distinct dendritic cell (DC) populations induced by physiologic stimuli: prostaglandin E2 regulates the migratory capacity of specific DC subsets.
Blood
100
:
1362
1372
.
63
Scandella
E.
,
Men
Y.
,
Gillessen
S.
,
Förster
R.
,
Groettrup
M.
.
2002
.
Prostaglandin E2 is a key factor for CCR7 surface expression and migration of monocyte-derived dendritic cells.
Blood
100
:
1354
1361
.
64
van Helden
S. F.
,
Krooshoop
D. J.
,
Broers
K. C.
,
Raymakers
R. A.
,
Figdor
C. G.
,
van Leeuwen
F. N.
.
2006
.
A critical role for prostaglandin E2 in podosome dissolution and induction of high-speed migration during dendritic cell maturation.
J. Immunol.
177
:
1567
1574
.
65
Yen
J. H.
,
Khayrullina
T.
,
Ganea
D.
.
2008
.
PGE2-induced metalloproteinase-9 is essential for dendritic cell migration.
Blood
111
:
260
270
.
66
Muthuswamy
R.
,
Mueller-Berghaus
J.
,
Haberkorn
U.
,
Reinhart
T. A.
,
Schadendorf
D.
,
Kalinski
P.
.
2010
.
PGE2 transiently enhances DC expression of CCR7 but inhibits the ability of DCs to produce CCL19 and attract naive T cells.
Blood
116
:
1454
1459
.
67
Baratelli
F. E.
,
Heuzé-Vourc’h
N.
,
Krysan
K.
,
Dohadwala
M.
,
Riedl
K.
,
Sharma
S.
,
Dubinett
S. M.
.
2004
.
Prostaglandin E2-dependent enhancement of tissue inhibitors of metalloproteinases-1 production limits dendritic cell migration through extracellular matrix.
J. Immunol.
173
:
5458
5466
.
68
De Vries
I. J.
,
Krooshoop
D. J.
,
Scharenborg
N. M.
,
Lesterhuis
W. J.
,
Diepstra
J. H.
,
Van Muijen
G. N.
,
Strijk
S. P.
,
Ruers
T. J.
,
Boerman
O. C.
,
Oyen
W. J.
, et al
.
2003
.
Effective migration of antigen-pulsed dendritic cells to lymph nodes in melanoma patients is determined by their maturation state.
Cancer Res.
63
:
12
17
.
69
Quillien
V.
,
Moisan
A.
,
Carsin
A.
,
Lesimple
T.
,
Lefeuvre
C.
,
Adamski
H.
,
Bertho
N.
,
Devillers
A.
,
Leberre
C.
,
Toujas
L.
.
2005
.
Biodistribution of radiolabelled human dendritic cells injected by various routes.
Eur. J. Nucl. Med. Mol. Imaging
32
:
731
741
.
70
Monrad
S. U.
,
Kojima
F.
,
Kapoor
M.
,
Kuan
E. L.
,
Sarkar
S.
,
Randolph
G. J.
,
Crofford
L. J.
.
2011
.
Genetic deletion of mPGES-1 abolishes PGE2 production in murine dendritic cells and alters the cytokine profile, but does not affect maturation or migration.
Prostaglandins Leukot. Essent. Fatty Acids
84
:
113
121
.
71
Doyen
V.
,
Rubio
M.
,
Braun
D.
,
Nakajima
T.
,
Abe
J.
,
Saito
H.
,
Delespesse
G.
,
Sarfati
M.
.
2003
.
Thrombospondin 1 is an autocrine negative regulator of human dendritic cell activation.
J. Exp. Med.
198
:
1277
1283
.
72
Braun
D.
,
Longman
R. S.
,
Albert
M. L.
.
2005
.
A two-step induction of indoleamine 2,3 dioxygenase (IDO) activity during dendritic-cell maturation.
Blood
106
:
2375
2381
.
73
Kaliński
P.
,
Hilkens
C. M.
,
Wierenga
E. A.
,
Kapsenberg
M. L.
.
1999
.
T-cell priming by type-1 and type-2 polarized dendritic cells: the concept of a third signal.
Immunol. Today
20
:
561
567
.
74
Gustafsson
K.
,
Ingelsten
M.
,
Bergqvist
L.
,
Nyström
J.
,
Andersson
B.
,
Karlsson-Parra
A.
.
2008
.
Recruitment and activation of natural killer cells in vitro by a human dendritic cell vaccine.
Cancer Res.
68
:
5965
5971
.
75
Hokey
D. A.
,
Larregina
A. T.
,
Erdos
G.
,
Watkins
S. C.
,
Falo
L. D.
 Jr.
2005
.
Tumor cell loaded type-1 polarized dendritic cells induce Th1-mediated tumor immunity.
Cancer Res.
65
:
10059
10067
.
76
Mailliard
R. B.
,
Wankowicz-Kalinska
A.
,
Cai
Q.
,
Wesa
A.
,
Hilkens
C. M.
,
Kapsenberg
M. L.
,
Kirkwood
J. M.
,
Storkus
W. J.
,
Kalinski
P.
.
2004
.
Alpha-type-1 polarized dendritic cells: a novel immunization tool with optimized CTL-inducing activity.
Cancer Res.
64
:
5934
5937
.
77
Campbell
D. J.
,
Kim
C. H.
,
Butcher
E. C.
.
2003
.
Chemokines in the systemic organization of immunity.
Immunol. Rev.
195
:
58
71
.
78
Salcedo
R.
,
Zhang
X.
,
Young
H. A.
,
Michael
N.
,
Wasserman
K.
,
Ma
W. H.
,
Martins-Green
M.
,
Murphy
W. J.
,
Oppenheim
J. J.
.
2003
.
Angiogenic effects of prostaglandin E2 are mediated by up-regulation of CXCR4 on human microvascular endothelial cells.
Blood
102
:
1966
1977
.
79
Kolenko
V.
,
Rayman
P.
,
Roy
B.
,
Cathcart
M. K.
,
O’Shea
J.
,
Tubbs
R.
,
Rybicki
L.
,
Bukowski
R.
,
Finke
J.
.
1999
.
Downregulation of JAK3 protein levels in T lymphocytes by prostaglandin E2 and other cyclic adenosine monophosphate-elevating agents: impact on interleukin-2 receptor signaling pathway.
Blood
93
:
2308
2318
.
80
Hahn
T.
,
Alvarez
I.
,
Kobie
J. J.
,
Ramanathapuram
L.
,
Dial
S.
,
Fulton
A.
,
Besselsen
D.
,
Walker
E.
,
Akporiaye
E. T.
.
2006
.
Short-term dietary administration of celecoxib enhances the efficacy of tumor lysate-pulsed dendritic cell vaccines in treating murine breast cancer.
Int. J. Cancer
118
:
2220
2231
.
81
Haas
A. R.
,
Sun
J.
,
Vachani
A.
,
Wallace
A. F.
,
Silverberg
M.
,
Kapoor
V.
,
Albelda
S. M.
.
2006
.
Cycloxygenase-2 inhibition augments the efficacy of a cancer vaccine.
Clin. Cancer Res.
12
:
214
222
.
82
Betz
M.
,
Fox
B. S.
.
1991
.
Prostaglandin E2 inhibits production of Th1 lymphokines but not of Th2 lymphokines.
J. Immunol.
146
:
108
113
.
83
Snijdewint
F. G.
,
Kaliński
P.
,
Wierenga
E. A.
,
Bos
J. D.
,
Kapsenberg
M. L.
.
1993
.
Prostaglandin E2 differentially modulates cytokine secretion profiles of human T helper lymphocytes.
J. Immunol.
150
:
5321
5329
.
84
van der Pouw Kraan
T. C.
,
Boeije
L. C.
,
Smeenk
R. J.
,
Wijdenes
J.
,
Aarden
L. A.
.
1995
.
Prostaglandin-E2 is a potent inhibitor of human interleukin 12 production.
J. Exp. Med.
181
:
775
779
.
85
Wu
C. Y.
,
Wang
K.
,
McDyer
J. F.
,
Seder
R. A.
.
1998
.
Prostaglandin E2 and dexamethasone inhibit IL-12 receptor expression and IL-12 responsiveness.
J. Immunol.
161
:
2723
2730
.
86
Kaliński
P.
,
Vieira
P. L.
,
Schuitemaker
J. H.
,
de Jong
E. C.
,
Kapsenberg
M. L.
.
2001
.
Prostaglandin E2 is a selective inducer of interleukin-12 p40 (IL-12p40) production and an inhibitor of bioactive IL-12p70 heterodimer.
Blood
97
:
3466
3469
.
87
Whittaker
D. S.
,
Bahjat
K. S.
,
Moldawer
L. L.
,
Clare-Salzler
M. J.
.
2000
.
Autoregulation of human monocyte-derived dendritic cell maturation and IL-12 production by cyclooxygenase-2-mediated prostanoid production.
J. Immunol.
165
:
4298
4304
.
88
Gillessen
S.
,
Carvajal
D.
,
Ling
P.
,
Podlaski
F. J.
,
Stremlo
D. L.
,
Familletti
P. C.
,
Gubler
U.
,
Presky
D. H.
,
Stern
A. S.
,
Gately
M. K.
.
1995
.
Mouse interleukin-12 (IL-12) p40 homodimer: a potent IL-12 antagonist.
Eur. J. Immunol.
25
:
200
206
.
89
Kapsenberg
M. L.
,
Hilkens
C. M.
,
Wierenga
E. A.
,
Kalinski
P.
.
1999
.
The paradigm of type 1 and type 2 antigen-presenting cells: implications for atopic allergy.
Clin. Exp. Allergy
29
(
Suppl. 2
):
33
36
.
90
Sheibanie
A. F.
,
Yen
J. H.
,
Khayrullina
T.
,
Emig
F.
,
Zhang
M.
,
Tuma
R.
,
Ganea
D.
.
2007
.
The proinflammatory effect of prostaglandin E2 in experimental inflammatory bowel disease is mediated through the IL-23→IL-17 axis.
J. Immunol.
178
:
8138
8147
.
91
Woolard
M. D.
,
Hensley
L. L.
,
Kawula
T. H.
,
Frelinger
J. A.
.
2008
.
Respiratory Francisella tularensis live vaccine strain infection induces Th17 cells and prostaglandin E2, which inhibits generation of γ interferon-positive T cells.
Infect. Immun.
76
:
2651
2659
.
92
Esaki
Y.
,
Li
Y.
,
Sakata
D.
,
Yao
C.
,
Segi-Nishida
E.
,
Matsuoka
T.
,
Fukuda
K.
,
Narumiya
S.
.
2010
.
Dual roles of PGE2-EP4 signaling in mouse experimental autoimmune encephalomyelitis.
Proc. Natl. Acad. Sci. USA
107
:
12233
12238
.
93
Boniface
K.
,
Bak-Jensen
K. S.
,
Li
Y.
,
Blumenschein
W. M.
,
McGeachy
M. J.
,
McClanahan
T. K.
,
McKenzie
B. S.
,
Kastelein
R. A.
,
Cua
D. J.
,
de Waal Malefyt
R.
.
2009
.
Prostaglandin E2 regulates Th17 cell differentiation and function through cyclic AMP and EP2/EP4 receptor signaling.
J. Exp. Med.
206
:
535
548
.
94
Khayrullina
T.
,
Yen
J. H.
,
Jing
H.
,
Ganea
D.
.
2008
.
In vitro differentiation of dendritic cells in the presence of prostaglandin E2 alters the IL-12/IL-23 balance and promotes differentiation of Th17 cells.
J. Immunol.
181
:
721
735
.
95
Hale
A. H.
,
Evans
D. L.
,
Daniel
L. W.
.
1982
.
Effect of prostaglandins on elicitation of anti-viral-cytolytic activity.
Immunol. Lett.
4
:
171
174
.
96
Plaut
M.
,
Marone
G.
,
Gillespie
E.
.
1983
.
The role of cyclic AMP in modulating cytotoxic T lymphocytes, II: Sequential changes during culture in responsiveness of cytotoxic lymphocytes to cyclic AMP-active agents.
J. Immunol.
131
:
2945
2952
.
97
Lala
P. K.
,
Kennedy
T. G.
,
Parhar
R. S.
.
1988
.
Suppression of lymphocyte alloreactivity by early gestational human decidua, II: Characterization of the suppressor mechanisms.
Cell. Immunol.
116
:
411
422
.
98
Parhar
R. S.
,
Lala
P. K.
.
1988
.
Prostaglandin E2-mediated inactivation of various killer lineage cells by tumor-bearing host macrophages.
J. Leukoc. Biol.
44
:
474
484
.
99
Specht
C.
,
Bexten
S.
,
Kölsch
E.
,
Pauels
H. G.
.
2001
.
Prostaglandins, but not tumor-derived IL-10, shut down concomitant tumor-specific CTL responses during murine plasmacytoma progression.
Int. J. Cancer
91
:
705
712
.
100
Valitutti
S.
,
Dessing
M.
,
Lanzavecchia
A.
.
1993
.
Role of cAMP in regulating cytotoxic T lymphocyte adhesion and motility.
Eur. J. Immunol.
23
:
790
795
.
101
Watchmaker
P. B.
,
Berk
E.
,
Muthuswamy
R.
,
Mailliard
R. B.
,
Urban
J. A.
,
Kirkwood
J. M.
,
Kalinski
P.
.
2010
.
Independent regulation of chemokine responsiveness and cytolytic function versus CD8+ T cell expansion by dendritic cells.
J. Immunol.
184
:
591
597
.
102
Garcia-Peñarrubia
P.
,
Bankhurst
A. D.
,
Koster
F. T.
.
1989
.
Prostaglandins from human T suppressor/cytotoxic cells modulate natural killer antibacterial activity.
J. Exp. Med.
170
:
601
606
.
103
Simkin
N. J.
,
Jelinek
D. F.
,
Lipsky
P. E.
.
1987
.
Inhibition of human B cell responsiveness by prostaglandin E2.
J. Immunol.
138
:
1074
1081
.
104
Carini
C.
,
Hudspith
B. N.
,
Brostoff
J.
.
1981
.
Effect of prostaglandins and cyclic nucleotides on growth and immunoglobulin secretion of two IgE myeloma cell lines.
Br. J. Cancer
43
:
257
260
.
105
Baratelli
F.
,
Lin
Y.
,
Zhu
L.
,
Yang
S. C.
,
Heuzé-Vourc’h
N.
,
Zeng
G.
,
Reckamp
K.
,
Dohadwala
M.
,
Sharma
S.
,
Dubinett
S. M.
.
2005
.
Prostaglandin E2 induces FOXP3 gene expression and T regulatory cell function in human CD4+ T cells.
J. Immunol.
175
:
1483
1490
.
106
Sharma
S.
,
Yang
S. C.
,
Zhu
L.
,
Reckamp
K.
,
Gardner
B.
,
Baratelli
F.
,
Huang
M.
,
Batra
R. K.
,
Dubinett
S. M.
.
2005
.
Tumor cyclooxygenase-2/prostaglandin E2-dependent promotion of FOXP3 expression and CD4+CD25+ T regulatory cell activities in lung cancer.
Cancer Res.
65
:
5211
5220
.
107
Bergmann
C.
,
Strauss
L.
,
Zeidler
R.
,
Lang
S.
,
Whiteside
T. L.
.
2007
.
Expansion of human T regulatory type 1 cells in the microenvironment of cyclooxygenase 2 overexpressing head and neck squamous cell carcinoma.
Cancer Res.
67
:
8865
8873
.
108
Soontrapa
K.
,
Honda
T.
,
Sakata
D.
,
Yao
C.
,
Hirata
T.
,
Hori
S.
,
Matsuoka
T.
,
Kita
Y.
,
Shimizu
T.
,
Kabashima
K.
,
Narumiya
S.
.
2011
.
Prostaglandin E2-prostaglandin E receptor subtype 4 (EP4) signaling mediates UV irradiation-induced systemic immunosuppression.
Proc. Natl. Acad. Sci. USA
108
:
6668
6673
.
109
Muthuswamy
R.
,
Urban
J.
,
Lee
J. J.
,
Reinhart
T. A.
,
Bartlett
D.
,
Kalinski
P.
.
2008
.
Ability of mature dendritic cells to interact with regulatory T cells is imprinted during maturation.
Cancer Res.
68
:
5972
5978
.
110
Banerjee
D. K.
,
Dhodapkar
M. V.
,
Matayeva
E.
,
Steinman
R. M.
,
Dhodapkar
K. M.
.
2006
.
Expansion of FOXP3high regulatory T cells by human dendritic cells (DCs) in vitro and after injection of cytokine-matured DCs in myeloma patients.
Blood
108
:
2655
2661
.
111
Mahic
M.
,
Yaqub
S.
,
Johansson
C. C.
,
Taskén
K.
,
Aandahl
E. M.
.
2006
.
FOXP3+CD4+CD25+ adaptive regulatory T cells express cyclooxygenase-2 and suppress effector T cells by a prostaglandin E2-dependent mechanism.
J. Immunol.
177
:
246
254
.
112
Huang
M.
,
Stolina
M.
,
Sharma
S.
,
Mao
J. T.
,
Zhu
L.
,
Miller
P. W.
,
Wollman
J.
,
Herschman
H.
,
Dubinett
S. M.
.
1998
.
Non-small cell lung cancer cyclooxygenase-2-dependent regulation of cytokine balance in lymphocytes and macrophages: up-regulation of interleukin 10 and down-regulation of interleukin 12 production.
Cancer Res.
58
:
1208
1216
.
113
Stolina
M.
,
Sharma
S.
,
Lin
Y.
,
Dohadwala
M.
,
Gardner
B.
,
Luo
J.
,
Zhu
L.
,
Kronenberg
M.
,
Miller
P. W.
,
Portanova
J.
, et al
.
2000
.
Specific inhibition of cyclooxygenase 2 restores antitumor reactivity by altering the balance of IL-10 and IL-12 synthesis.
J. Immunol.
164
:
361
370
.
114
Ochoa
A. C.
,
Zea
A. H.
,
Hernandez
C.
,
Rodriguez
P. C.
.
2007
.
Arginase, prostaglandins, and myeloid-derived suppressor cells in renal cell carcinoma.
Clin. Cancer Res.
13
:
721s
726s
.
115
Sinha
P.
,
Clements
V. K.
,
Fulton
A. M.
,
Ostrand-Rosenberg
S.
.
2007
.
Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells.
Cancer Res.
67
:
4507
4513
.
116
Fujita
M.
,
Kohanbash
G.
,
Fellows-Mayle
W.
,
Hamilton
R. L.
,
Komohara
Y.
,
Decker
S. A.
,
Ohlfest
J. R.
,
Okada
H.
.
2011
.
COX-2 blockade suppresses gliomagenesis by inhibiting myeloid-derived suppressor cells.
Cancer Res.
71
:
2664
2674
.
117
Serafini
P.
2010
.
Editorial: PGE2-producing MDSC: a role in tumor progression?
J. Leukoc. Biol.
88
:
827
829
.
118
Jung
Y. J.
,
Isaacs
J. S.
,
Lee
S.
,
Trepel
J.
,
Neckers
L.
.
2003
.
IL-1beta-mediated up-regulation of HIF-1α via an NFκB/COX-2 pathway identifies HIF-1 as a critical link between inflammation and oncogenesis.
FASEB J.
17
:
2115
2117
.
119
Corzo
C. A.
,
Condamine
T.
,
Lu
L.
,
Cotter
M. J.
,
Youn
J. I.
,
Cheng
P.
,
Cho
H. I.
,
Celis
E.
,
Quiceno
D. G.
,
Padhya
T.
, et al
.
2010
.
HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment.
J. Exp. Med.
207
:
2439
2453
.
120
Freire-de-Lima
C. G.
,
Nascimento
D. O.
,
Soares
M. B.
,
Bozza
P. T.
,
Castro-Faria-Neto
H. C.
,
de Mello
F. G.
,
DosReis
G. A.
,
Lopes
M. F.
.
2000
.
Uptake of apoptotic cells drives the growth of a pathogenic trypanosome in macrophages.
Nature
403
:
199
203
.
121
Mittal
R.
,
Gonzalez-Gomez
I.
,
Panigrahy
A.
,
Goth
K.
,
Bonnet
R.
,
Prasadarao
N. V.
.
2010
.
IL-10 administration reduces PGE-2 levels and promotes CR3-mediated clearance of Escherichia coli K1 by phagocytes in meningitis.
J. Exp. Med.
207
:
1307
1319
.
122
Zeidler
R.
,
Csanady
M.
,
Gires
O.
,
Lang
S.
,
Schmitt
B.
,
Wollenberg
B.
.
2000
.
Tumor cell-derived prostaglandin E2 inhibits monocyte function by interfering with CCR5 and Mac-1.
FASEB J.
14
:
661
668
.
123
Van Elssen
C. H.
,
Vanderlocht
J.
,
Oth
T.
,
Senden-Gijsbers
B. L.
,
Germeraad
W. T.
,
Bos
G. M.
.
2011
.
Inflammation restraining effects of prostaglandin E2 on NK-DC interaction are imprinted during dendritic cell maturation.
Blood
118
:
2473
2482
.
124
McIlroy
A.
,
Caron
G.
,
Blanchard
S.
,
Frémaux
I.
,
Duluc
D.
,
Delneste
Y.
,
Chevailler
A.
,
Jeannin
P.
.
2006
.
Histamine and prostaglandin E up-regulate the production of Th2-attracting chemokines (CCL17 and CCL22) and down-regulate IFN-γ-induced CXCL10 production by immature human dendritic cells.
Immunology
117
:
507
516
.
125
Obermajer
N.
,
Muthuswamy
R.
,
Odunsi
K. O.
,
Edwards
R. P.
,
Kalinski
P.
.
2011
.
PGE2-dependent CXCL12 production and CXCR4 expression control the accumulation of human MDSCs in ovarian cancer environment.
Cancer Res.
DOI: 10.1158/0008-5472.CAN-11-2449
.
126
Stock
A.
,
Booth
S.
,
Cerundolo
V.
.
2011
.
Prostaglandin E2 suppresses the differentiation of retinoic acid-producing dendritic cells in mice and humans.
J. Exp. Med.
208
:
761
773
.
127
Oppenheimer-Marks
N.
,
Kavanaugh
A. F.
,
Lipsky
P. E.
.
1994
.
Inhibition of the transendothelial migration of human T lymphocytes by prostaglandin E2.
J. Immunol.
152
:
5703
5713
.
128
Mesri
M.
,
Liversidge
J.
,
Forrester
J. V.
.
1996
.
Prostaglandin E2 and monoclonal antibody to lymphocyte function-associated antigen-1 differentially inhibit migration of T lymphocytes across microvascular retinal endothelial cells in rat.
Immunology
88
:
471
477
.
129
Vancheri
C.
,
Mastruzzo
C.
,
Sortino
M. A.
,
Crimi
N.
.
2004
.
The lung as a privileged site for the beneficial actions of PGE2.
Trends Immunol.
25
:
40
46
.

The author has no financial conflicts of interest.