Skip to main content

Crosstalk between purinergic receptors and lipid mediators in leishmaniasis

Abstract

Leishmaniasis is a neglected tropical disease affecting millions of people around the world caused by organisms of the genus Leishmania. Parasite escape mechanisms of the immune system confer the possibility of resistance and dissemination of the disease. A group of molecules that has become a target for Leishmania survival strategies are lipid mediators. Among them, leukotriene B4 (LTB4) has been described as a pro-inflammatory molecule capable of activating cells of the immune system to combat Leishmania. In an opposite way, prostaglandin E2 (PGE2) is a lipid mediator described as a deactivator of macrophages and neutrophils. The balance of these two molecules can be generated by extracellular nucleotides, such as adenosine 5'-triphosphate (ATP) and adenosine (Ado), which activate the purinergic receptors system. Herein, we discuss the role of extracellular nucleotides and the resulting balance of LTB4 and PGE2 in Leishmania fate, survival or death.

Review

Background

The protozoan parasites of the genus Leishmania cause a broad range of human diseases called leishmaniasis. Leishmaniasis is a neglected disease of tropical and subtropical areas that affects more than 12 million people worldwide [1]. Moreover, every year, 2 million new cases are diagnosed, among them, 75 % of the cases are cutaneous and 25 % are visceral leishmaniasis, leading to it being the second most common cause of parasite-associated death resulting in 20,000 to 30,000 deaths per year [2]. Leishmania preferentially infect phagocytic cells, as macrophages, neutrophils and dendritic cells of susceptible mammalian hosts [3] causing numerous clinical manifestations. In general, cutaneous leishmaniasis is located adjacent to the infectious site, the skin or lymph nodes. The parasite can escape into the nasal and oropharyngeal mucosa causing mucocutaneous leishmaniasis; or also migrate to the spleen, liver, bone marrow, and distant lymph nodes, leading to lethal clinical manifestations, named visceral leishmaniasis or kala-azar [4].

Leishmaniasis is transmitted by a female blood-sucking insects of the genus Phlebotomus in the 'Old' World and by species of Lutzomya in the 'New' World. The parasite can occur in two ways: the promastigote, which has high mobility, and is found in the digestive tract of the vector; and amastigote, without flagella, which develops into the phagolysosomes of phagocytic cells. Immediately before blood intake, the insect saliva containing promastigote forms is inoculated into the skin of the mammalian host. Soon after, the parasite is phagocytosed, remaining viable inside the phagolysosome, the fused phagosome and lysosome. Then, the promastigote form differentiates in amastigote approximately 12–24 h later [5, 6]. When an infected mammal host is bitten by the sand fly, it sucks amastigote-infected macrophages or free amastigotes which will transform into mobile flagellated promastigotes in the midgut of the vector. In this process, procyclic promastigotes (proliferative and non-infective forms) acquire the ability to be virulent and non-proliferative, the metacyclic promastigotes, and this process is called metacyclogenesis [7]. These promastigote forms migrate to the oral cavity promoting the transmission in the next blood meal.

The innate immune cells present in the skin are the first line of defense against Leishmania infection [8]. Dermal dendritic cells (DCs), Langerhans cells (LCs) [9, 10], mast cells, T cells, and macrophages are the immune cells in the skin. Interesting, keratinocytes, which are the most abundant in the skin, also play an active role in the local immune response and it has been reported that they have an important role in polarization of the Th1 response during leishmaniasis [11, 12]. After parasite inoculum into the dermis, neutrophils quickly infiltrate and phagocytose Leishmania parasites, becoming the first circulatory cells to reach tissue space [13–15]. Macrophages are the second wave of infiltrating immune cells and are the principal host cells for the Leishmania [16, 17]. Thus, neutrophils and macrophages play crucial roles in disease progression, but ironically as professional phagocytic and killing cells, they become targets because of evasion mechanisms employed by Leishmania to subvert the host immune system.

Macrophages and neutrophils possess several pattern recognition receptors (PRR) that respond to pathogen-associated molecular patterns (PAMPs) present in the Leishmania surface, such as lipophosphoglycan (LPG) and glycoprotein 63 (GP63), both in humans and in mice [18–21]. Several host immune receptors can bind Leishmania components or antibodies against Leishmania, such as: the first and third complement receptor (CR1 and CR3, respectively) [22, 23], mannose receptor (MR) [24, 25], Fc gamma receptors (FcγRs) [26], fibronectin receptors (FNRS) [18], and Toll-like receptors (TLR) [27–30]. TLRs are phylogenetically the most ancient and best studied PRRs. In humans, 10 TLR family members have been identified and there are 12 in mice [31]. TLRs activation has been associated with the production and release of inflammatory mediators, such as cytokines, lipid mediators, and adenosine 5’-triphosphate (ATP) in extracellular medium [32–34]. ATP is widely present in the intracellular environment, at concentrations in the millimolar range, whereas it is almost imperceptible in the extracellular space, being around nanomolar [35]. We recently showed that Leishmania amazonensis recognition by macrophages leads to ATP release [36] (Fig. 1a). However, the receptor involved in this release and the mechanism that triggers this process has not yet been elucidated.

Fig. 1
figure 1

Schematic representation of elimination and evasion mechanisms mediated by purinergic signaling and lipid mediators during Leishmania infection. a Leishmania spp. promastigotes can be recognized by PRRs. This recognition leads to the release of ATP into the extracellular medium. b eATP active P2X7 receptors, which in turn leads to release of LTB4. c LTB4 binds to specific receptors on cell membrane, as BLT1, causing the elimination of Leishmania spp. by production of ROS, NO, and participates on Th1 and Th17 polarization. d In order to evade the immune system and ensure its survival, Leishmania spp. possess ecto-nucleotidase enzymes, such as E-NTPDase and ecto-5’-nucleotidase, removing eATP and favoring Ado accumulation. e Ado actives P1 receptors, such as A2B, increasing COX-2 expression and therefore leads to the release of PGE2. f PGE2 in turn binds EP receptors on cellular membranes, causing the decrease of ROS and NO production, and participates on Th2 polarization, resulting in establishment and dissemination of Leishmania spp. infection

Purinergic receptors

Extracellular ATP (eATP) is defined as a damage-associated molecular pattern (DAMP) causing biological effects though the activation of purinergic receptors that are presented on the cell membrane. Purinergic receptors are classified into two large families: P1 and P2. The P1 receptor family is characterized by metabotropic receptors activated by adenosine: A1, A2a, A2b, and A3 [37]. The P1 receptor activation has been discussed in several systems, suggesting a role in both physiological and pathological processes. In the immune system, P1 receptors are widely expressed by cells of the myeloid and lymphoid lineage [38]. P1 receptors act in regulating the immune response, and are involved mainly in resolving inflammation [39].

The P2 family of receptors is subdivided into P2X and P2Y. The P2Y receptors are G-protein coupled receptors, [35] while P2X are ionotropic receptors, capable of forming cationic channels activated by eATP. The participation of P2Y receptors in inflammatory events has been described [40, 41]. Furthermore, it has been reported that P2Y2 receptors act in neutrophil chemotaxis after activation by eATP [42]. The family of P2X receptors, in contrast, consists of ionotropic receptors. These receptors are intrinsic ion channels for Na+, K+, and Ca2+. To date, seven subtypes of P2X family have been cloned: P2X1 to P2X7 [43, 44]. The most studied of P2X receptors is the P2X7 subtype. This receptor has two transmembrane domains, being a polypeptide of 595 amino acids with a longer C-terminal domain, compared to other members of the P2X family. This peculiarity makes it capable of inducing the formation of pores permeable to molecules up to 900 Daltons after sustained eATP stimulation [45]. Moreover, the elongated C-terminal enables it to initiate various intracellular signaling cascades culminating with apoptosis, vesicular fusion, phospolipase D activation, exosome release, activation and secretion of pro-inflammatory cytokines IL-1β and IL-18 [44]. The expression of P2X7 receptor is well characterized under many cell types, including macrophages [46], monocytes [47], neutrophils [48], among others [49]. Furthermore, many studies have demonstrated the participation of purinergic receptors in the induction of bioactive lipid mediators [50–54].

Lipid mediators

Lipid metabolites of arachidonic acid (AA), including leukotrienes (LTs) and prostaglandins (PGs), have emerged as important mediators of a variety of physiological and pathophysiological functions. They are synthesized through the metabolism of AA released by cytosolic phospholipase A2. The lipid metabolites can be subsequently metabolized by different pathways, including the cycloxygenase (COX) enzymes and lipoxygenase (LO) generating a range of bioactive eicosanoids, named PGs and LTs, respectively. The activation of cPLA2 and 5-LO involves an increase of intracellular Ca2+ and subsequently activation of certain protein kinases, as well as translocation of 5-LO from cytoplasm and nucleoplasm to membrane sites such as the nuclear envelope [55]. The AA is presented to 5-LO by an essential accessory protein called 5-LO activating protein (FLAP), producing an unstable precursor of all other leukotrienes, the LTA4 [55]. Once generated, LTA4 can be conjugated with reduced glutathione by LTC4 synthase (LTC4S) to form LTC4, or LTA4 can also be hydrolyzed by LTA4 hydrolase (LTA4H) to form LTB4 [56]. LTC4 as LTB4 can be exported to the extracellular space through specific transporters [57–59]. In the extracellular environment, LTC4 is rapidly converted to LTD4 by the glutamyl leukotrienase removing glutamic acid molecule of LTC4, and LTD4 can be further converted to LTE4 by the dipeptidase which removes a glycine residue of LTD4 molecule [60]. LTB4 is best known as a chemotactic and activator for leukocytes, and cysteinyl leukotrienes (LTC4, LTD4, and LTE4) are widely known in the pathogenesis of asthma [61].

PGs are formed when AA is metabolized by sequential actions of cyclooxygenase and their specific synthases [62]. COX has both cyclooxygenase (COX) and peroxidase activity, and three COX isoforms were described: COX-1, COX-2 and COX-3 [63, 64]. COX-1 and COX-3 are constitutively expressed while COX-2 is induced by inflammatory stimuli [64, 65]. There are six bioactive PGs: PGE2, PGI2, PGD2 and PGF2 [62]. Much is known about the pro-inflammatory functions of PGs, but, in the past years, it has been proven to also possess potential anti-inflammatory effects of PGs observed in resolution phase [66], and, importantly, these effects can be used by parasites to evade the immune system.

Purinergic receptor, lipid mediators and immune evasion

The most effective mechanisms against infection by Leishmania already described involve the production of reactive oxygen species (ROS) and nitric oxide (NO) [67]. Furthermore, it has been shown that an effective response against infection by Leishmania is given by the induction of Th1 and Th17 responses [68–72], while Th2 response promotes susceptibility [68, 70].

The role of extracellular nucleotides and the activation of purinergic receptors during infection by L. amazonensis have been investigated [73]. Marques-da-Silva and colleagues [74] showed that P2Y2 and P2Y4 receptors have its expression upregulated and increased levels of uridine triphosphate (UTP) nucleotide into the extracellular environment during infection can lead to death of the macrophage by apoptosis and the elimination of the parasite. Other studies have shown that eATP can lead to the elimination of L. amazonensis in infected macrophages via P2X7 receptor [75]. A recent study demonstrates that elimination of L. amazonensis by P2X7 receptor depends on the production of LTB4 and leukotriene B4 receptor 1 (BLT1) [36] (Fig. 1b, c). Additionally, other studies have demonstrated the production of LTB4 in resistance to L. amazonensis and L. braziliensis, in humans and mice [76–78]. Furthermore, latest studies have demonstrated the participation of 15d-Prostaglandin J2 in L. donovani elimination [79]. This resistance can be due to the production of ROS and NO, which may be produced after P2X7 receptor activation [80, 81] and LTB4 release [82–85]. Moreover, the P2X7 receptor activation and LTB4 release have been implicated in the polarization of Th1 and Th17 responses, participating in the immune response against Leishmania [86–90] (Fig. 1c).

Regarding the participation of lipid mediators in Leishmania infection, the role of PGE2 in susceptibility has been discussed. It is known that PGE2 possesses anti-inflammatory activity, facilitating Leishmania infection in macrophages, suppressing inflammatory response in both cutaneous and visceral leishmaniasis [91–94]. Moreover, reinforcing the context of a beneficial effect of PGE2 for Leishmania survival, it was demonstrated that several Leishmania species possess lipid corpuscles as organelles and L. infantum is able to produce and release PGs, such as PGF2α itself [95, 96] (Fig. 1d). It is important to highlight that PGE2 inhibits NO production [97], and Th1 and Th17 development [98–101] and, consequently, stimulates Th2 response, favoring infection [99] (Fig. 1f).

On the other hand, in order to perpetuate itself, Leishmania has developed methods to subvert microbicidal mechanisms and immune responses against itself. As already described before, eATP has proved to be an endogenous molecule able to induce the death of L. amazonensis through P2X7 receptors activation [36]. It has also been well established that the presence of enzymes capable of degrading ATP in the mammalian cell membrane forming ADP (adenosine-diphosphate) and adenosine (Ado), named ecto-nucleotidases. Among them, CD39 (ecto-NPTDase) and CD73 (ecto-5′-nucleotidase) exert relevant actions, regulating inflammatory responses of ATP and UTP. Thus, Ado is formed through the action of CD39 that converts ATP and ADP to 5'-adenosine mono-phosphate (AMP). AMP is the substrate for CD73. This enzyme, in turn, catalyzes the reaction that converts AMP to Ado [39, 102]. In this scenario, it has been shown that Leishmania express ecto-nucleotidase activity. This is confirmed by the observation of increased Ado levels in serum from visceral leishmaniasis patients [103, 104]. This can cause the prevention of the activation of macrophages and leads to the increase of infection by Leishmania species [105–109]. Moreover, the virulence of L. amazonensis promastigotes could be due to its high ecto-nucleotidase activity [110] (Fig. 1d). Moreover, ecto-5-nucleotidase activity also has been seen in L. chagasi [105]. Furthermore, it has been observed that L. amazonensis infection increases ecto-nucleotidases expression in DC [111]. Thus, the blocking of the A2B receptors is found to increase production of NO and decrease parasite survival, suggesting participation of Ado in this process [109].

Others have shown that Ado increases COX-2 expression and PGE2 production in neutrophils [112, 113] (Fig. 1e). This corroborates the fact that both Ado and PGE2 stimulates the release of anti-inflammatory cytokines, such as interleukin (IL)-10 in macrophages [114, 115], while inhibiting the release of pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-α and IL-12 in DCs and macrophages [116, 117]. This stimulates an anti-inflammatory environment, allowing establishment of infection.

It has been shown that Ado decreases production and release of LTB4 [118–121], which modulates microbicidal mechanisms. Moreover, it is known that L. amazonensis is capable to negatively modulate the production of LTB4 via P2X7 receptor activation in macrophages from C57BL/6 and BALB/c mice [36]. Neutrophils are recruited to the infection site when infection is initiated by sand fly bite [14, 122], spreading Leishmania parasites [17, 123, 124]. However, in other species of Leishmania, such as L. braziliensis, the neutrophils are important for parasite elimination [125]. Nevertheless, the role of the Ado in stimulation of PGE2 production in macrophages still needs to be studied. Moreover, other pathogens use Ado to subvert the immune system such as Toxoplasma gondii, Staphylococcus aureus and Streptococcus agalactiae [126–128].

The role of sand fly saliva substances in modulating Leishmania infection has been demonstrated [129, 130]. Furthermore, it has been described that sand fly saliva can inhibit NO production, and consequently increase the parasite load [131, 132]. It has also been described that Lutzomyia longipalpis saliva possesses ATPase activity, which can hydrolyse eATP [133]. Sand fly saliva also contains high levels of Ado, modulating the inflammatory micro-environment, causing NO inhibition, and macrophage inactivation, which in turn increases the parasitic load in macrophages and neutrophils [134–136]. Recently it was shown that exosomes are co-inoculated with Leishmania into mammalian hosts [137]. It is tempting to correlate it with a burst of ATP secretion, local Ado generation and PGE2 production. It is known that L. longipalpis saliva triggers the production and release of PGE2 and decreases LTB4 in macrophages [138, 139].

Conclusion

The establishment of Leishmania infection can be due to the balance of several factors. Extracellular nucleotides can modulate the balance of pro- and anti-inflammatory factors such as PGs and LTs. To ensure their survival, Leishmania spp. developed strategies throughout its evolution to guarantee its perpetuation (Fig. 2a). The ability of Leishmania spp. to modulate extracellular concentrations of ATP and Ado, and consequently the balance of LTB4 and PGE2 shows how organisms can subvert the immune system of the host (Fig. 2b). Thus, the importance of knowledge of these strategies of evasion is essential in order to develop drugs capable to counterbalance Leishmania evasion.

Fig. 2
figure 2

Balance between nucleotides and inflammatory lipid mediators on Leishmania spp. infection establishment. a The increase of ATP and reduction of Ado in extracellular medium leads to an overproduction of LTB4, which in turn stimulates the production of ROS and NO, and the polarization of immune responses for a Th1 and Th17 pattern; meanwhile a high PGE2 production also directs the polarization for Th2 response. This scenario facilitates the elimination of Leishmania spp. by macrophages. b The evasion of Leishmania spp. occurs when Ado concentrations in the extracellular medium overlaps the ATP. Thus, there is an increase in PGE2 and decreased ROS and NO, with consequent polarization Th2

Abbreviations

ATP:

5′-adenosine triphosphate

eATP:

extracellular ATP

Ado:

Adenosine

DC:

Dermal dendritic cells

LCs:

Langerhans cells

LPG:

Lipophosphoglyca

GP63:

Glycoprotein 63

CR:

Complement receptor

MR:

Mannose receptor

FcγRs:

Fc gamma receptors

FNRS:

Fibronectin receptors

TLR:

Toll-like receptor

DAMP:

Damage-associated molecular pattern; cytosolic phospholipase A2

COX:

Cicloxygenase

5-LO:

5-lipoxygenase

LTB4 :

Leukotriene B4

NO:

Nitric oxide

ROS:

Reactive oxygen species

PGE2 :

Prostaglandin E2

AMP:

5′-adenosine mono-phosphate

TNF:

Tumor necrosis factor

UTP:

Uridine triphosphate

PAMPs:

Pathogen-associated molecular patterns

References

  1. Desjeux P. Leishmaniasis: current situation and new perspectives. Comp Immunol Microbiol Infect Dis. 2004;27(5):305–18.

    Article  CAS  PubMed  Google Scholar 

  2. Alvar J, Velez ID, Bern C, Herrero M, Desjeux P, Cano J, et al. Leishmaniasis worldwide and global estimates of its incidence. PLoS One. 2012;7(5):e35671.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. van GJ, Diro E. Visceral leishmaniasis. Infect Dis Clin North Am. 2012;26(2):309–22.

    Article  Google Scholar 

  4. Murray HW, Berman JD, Davies CR, Saravia NG. Advances in leishmaniasis. Lancet. 2005;366(9496):1561–77.

    Article  CAS  PubMed  Google Scholar 

  5. Courret N, Frehel C, Prina E, Lang T, Antoine JC. Kinetics of the intracellular differentiation of Leishmania amazonensis and internalization of host MHC molecules by the intermediate parasite stages. Parasitology. 2001;122(Pt 3):263–79.

    Article  CAS  PubMed  Google Scholar 

  6. Galvao-Quintao L, Alfieri SC, Ryter A, Rabinovitch M. Intracellular differentiation of Leishmania amazonensis promastigotes to amastigotes: presence of megasomes, cysteine proteinase activity and susceptibility to leucine-methyl ester. Parasitology. 1990;101(Pt 1):7–13.

    Article  CAS  PubMed  Google Scholar 

  7. da Silva R, Sacks DL. Metacyclogenesis is a major determinant of Leishmania promastigote virulence and attenuation. Infect Immun. 1987;55(11):2802–6.

    PubMed  PubMed Central  Google Scholar 

  8. Solbach W, Laskay T. The host response to Leishmania infection. Adv Immunol. 2000;74:275–317.

    Article  CAS  PubMed  Google Scholar 

  9. Merad M, Ginhoux F, Collin M. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat Rev Immunol. 2008;8(12):935–47.

    Article  CAS  PubMed  Google Scholar 

  10. Nestle FO, Di MP, Qin JZ, Nickoloff BJ. Skin immune sentinels in health and disease. Nat Rev Immunol. 2009;9(10):679–91.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Albanesi C, Scarponi C, Giustizieri ML, Girolomoni G. Keratinocytes in inflammatory skin diseases. Curr Drug Targets Inflamm Allergy. 2005;4(3):329–34.

    Article  CAS  PubMed  Google Scholar 

  12. Ehrchen JM, Roebrock K, Foell D, Nippe N, von SE, Weiss JM, et al. Keratinocytes determine Th1 immunity during early experimental leishmaniasis. PLoS Pathog. 2010;6(4):e1000871.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Beil WJ, Meinardus-Hager G, Neugebauer DC, Sorg C. Differences in the onset of the inflammatory response to cutaneous leishmaniasis in resistant and susceptible mice. J Leukoc Biol. 1992;52(2):135–42.

    CAS  PubMed  Google Scholar 

  14. Peters NC, Egen JG, Secundino N, Debrabant A, Kimblin N, Kamhawi S, et al. In vivo imaging reveals an essential role for neutrophils in leishmaniasis transmitted by sand flies. Science. 2008;321(5891):970–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ribeiro-Gomes FL, Sacks D. The influence of early neutrophil-Leishmania interactions on the host immune response to infection. Front Cell Infect Microbiol. 2012;2:59.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Ribeiro-Gomes FL, Otero AC, Gomes NA, Moniz-De-Souza MC, Cysne-Finkelstein L, Arnholdt AC, et al. Macrophage interactions with neutrophils regulate Leishmania major infection. J Immunol. 2004;172(7):4454–62.

    Article  CAS  PubMed  Google Scholar 

  17. van ZG, Klinger M, Mueller A, Dannenberg S, Gebert A, Solbach W, et al. Cutting edge: neutrophil granulocyte serves as a vector for Leishmania entry into macrophages. J Immunol. 2004;173(11):6521–5.

    Article  Google Scholar 

  18. Brittingham A, Chen G, McGwire BS, Chang KP, Mosser DM. Interaction of Leishmania gp63 with cellular receptors for fibronectin. Infect Immun. 1999;67(9):4477–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Vargas-Inc, Tai W, Xin L, Hogg AE, Corry DB, Soong L. Distinct roles for MyD88 and Toll-like receptor 2 during Leishmania braziliensis infection in mice. Infect Immun. 2009;77(7):2948–56.

    Article  CAS  Google Scholar 

  20. Isnard A, Shio MT, Olivier M. Impact of Leishmania metalloprotease GP63 on macrophage signaling. Front Cell Infect Microbiol. 2012;2:72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Spath GF, Garraway LA, Turco SJ, Beverley SM. The role(s) of lipophosphoglycan (LPG) in the establishment of Leishmania major infections in mammalian hosts. Proc Natl Acad Sci U S A. 2003;100(16):9536–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Peters C, Aebischer T, Stierhof YD, Fuchs M, Overath P. The role of macrophage receptors in adhesion and uptake of Leishmania mexicana amastigotes. J Cell Sci. 1995;108(Pt 12):3715–24.

    CAS  PubMed  Google Scholar 

  23. Kedzierski L, Montgomery J, Bullen D, Curtis J, Gardiner E, Jimenez-Ruiz A, et al. A leucine-rich repeat motif of Leishmania parasite surface antigen 2 binds to macrophages through the complement receptor 3. J Immunol. 2004;172(8):4902–6.

    Article  CAS  PubMed  Google Scholar 

  24. Blackwell JM, Ezekowitz RA, Roberts MB, Channon JY, Sim RB, Gordon S. Macrophage complement and lectin-like receptors bind Leishmania in the absence of serum. J Exp Med. 1985;162(1):324–31.

    Article  CAS  PubMed  Google Scholar 

  25. Akilov OE, Kasuboski RE, Carter CR, McDowell MA. The role of mannose receptor during experimental leishmaniasis. J Leukoc Biol. 2007;81(5):1188–96.

    Article  CAS  PubMed  Google Scholar 

  26. Kima PE, Constant SL, Hannum L, Colmenares M, Lee KS, Haberman AM, et al. Internalization of Leishmania mexicana complex amastigotes via the Fc receptor is required to sustain infection in murine cutaneous leishmaniasis. J Exp Med. 2000;191(6):1063–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kropf P, Freudenberg N, Kalis C, Modolell M, Herath S, Galanos C, et al. Infection of C57BL/10ScCr and C57BL/10ScNCr mice with Leishmania major reveals a role for Toll-like receptor 4 in the control of parasite replication. J Leukoc Biol. 2004;76(1):48–57.

    Article  CAS  PubMed  Google Scholar 

  28. Whitaker SM, Colmenares M, Pestana KG, McMahon-Pratt D. Leishmania pifanoi proteoglycolipid complex P8 induces macrophage cytokine production through Toll-like receptor 4. Infect Immun. 2008;76(5):2149–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cezario GA, de Oliveira LR, Peresi E, Nicolete VC, Polettini J, de Lima CR, et al. Analysis of the expression of toll-like receptors 2 and 4 and cytokine production during experimental Leishmania chagasi infection. Mem Inst Oswaldo Cruz. 2011;106(5):573–83.

    Article  CAS  PubMed  Google Scholar 

  30. Srivastava S, Pandey SP, Jha MK, Chandel HS, Saha B. Leishmania expressed lipophosphoglycan interacts with Toll-like receptor (TLR)-2 to decrease TLR-9 expression and reduce anti-leishmanial responses. Clin Exp Immunol. 2013;172(3):403–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11(5):373–84.

    Article  CAS  PubMed  Google Scholar 

  32. Cohen M, Volpin G, Meir T, Klein E, Katz R, Assaf M, et al. Possible association of Toll-like receptor 9 polymorphisms with cytokine levels and posttraumatic symptoms in individuals with various types of orthopaedic trauma: early findings. Injury. 2013;44(11):1625–9.

    Article  PubMed  Google Scholar 

  33. Park JA, Kim Y. Eicosanoid biosynthesis is activated via Toll, but not Imd signal pathway in response to fungal infection. J Invertebr Pathol. 2012;110(3):382–8.

    Article  CAS  PubMed  Google Scholar 

  34. Ren H, Teng Y, Tan B, Zhang X, Jiang W, Liu M, et al. Toll-like receptor-triggered calcium mobilization protects mice against bacterial infection through extracellular ATP release. Infect Immun. 2014;82(12):5076–85.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Burnstock G. Physiology and pathophysiology of purinergic neurotransmission. Physiol Rev. 2007;87(2):659–797.

    Article  CAS  PubMed  Google Scholar 

  36. Chaves MM, Marques-da-Silva C, Monteiro AP, Canetti C, Coutinho-Silva R. Leukotriene B4 modulates P2X7 receptor-mediated Leishmania amazonensis elimination in murine macrophages. J Immunol. 2014;192(10):4765–73.

    Article  CAS  PubMed  Google Scholar 

  37. Fredholm BB, IJzerman AP, Jacobson KA, Klotz KN, Linden J. International Union of Pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacol Rev. 2001;53(4):527–52.

    CAS  PubMed  Google Scholar 

  38. Di VF, Vuerich M. Purinergic signaling in the immune system. Auton Neurosci. 2015;191:117–23.

    Article  CAS  Google Scholar 

  39. Hasko G, Linden J, Cronstein B, Pacher P. Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug Discov. 2008;7(9):759–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Santiago-Perez LI, Flores RV, Santos-Berrios C, Chorna NE, Krugh B, Garrad RC, et al. P2Y(2) nucleotide receptor signaling in human monocytic cells: activation, desensitization and coupling to mitogen-activated protein kinases. J Cell Physiol. 2001;187(2):196–208.

    Article  CAS  PubMed  Google Scholar 

  41. Brunschweiger A, Muller CE. P2 receptors activated by uracil nucleotides - an update. Curr Med Chem. 2006;13(3):289–312.

    Article  CAS  PubMed  Google Scholar 

  42. Chen Y, Corriden R, Inoue Y, Yip L, Hashiguchi N, Zinkernagel A, et al. ATP release guides neutrophil chemotaxis via P2Y2 and A3 receptors. Science. 2006;314(5806):1792–5.

    Article  CAS  PubMed  Google Scholar 

  43. Abbracchio MP, Burnstock G, Verkhratsky A, Zimmermann H. Purinergic signalling in the nervous system: an overview. Trends Neurosci. 2009;32(1):19–29.

    Article  CAS  PubMed  Google Scholar 

  44. North RA. Molecular physiology of P2X receptors. Physiol Rev. 2002;82(4):1013–67.

    Article  CAS  PubMed  Google Scholar 

  45. Nihei OK, Savino W, Alves LA. Procedures to characterize and study P2Z/P2X7 purinoceptor: flow cytometry as a promising practical, reliable tool. Mem Inst Oswaldo Cruz. 2000;95(3):415–28.

    Article  CAS  PubMed  Google Scholar 

  46. Surprenant A, Rassendren F, Kawashima E, North RA, Buell G. The cytolytic P2Z receptor for extracellular ATP identified as a P2X receptor (P2X7). Science. 1996;272(5262):735–8.

    Article  CAS  PubMed  Google Scholar 

  47. Humphreys BD, Dubyak GR. Modulation of P2X7 nucleotide receptor expression by pro- and anti-inflammatory stimuli in THP-1 monocytes. J Leukoc Biol. 1998;64(2):265–73.

    CAS  PubMed  Google Scholar 

  48. Suh BC, Kim JS, Namgung U, Ha H, Kim KT. P2X7 nucleotide receptor mediation of membrane pore formation and superoxide generation in human promyelocytes and neutrophils. J Immunol. 2001;166(11):6754–63.

    Article  CAS  PubMed  Google Scholar 

  49. Burnstock G, Boeynaems JM. Purinergic signalling and immune cells. Purinergic Signal. 2014;10(4):529–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Alzola E, Perez-Etxebarria A, Kabre E, Fogarty DJ, Metioui M, Chaib N, et al. Activation by P2X7 agonists of two phospholipases A2 (PLA2) in ductal cells of rat submandibular gland. Coupling of the calcium-independent PLA2 with kallikrein secretion. J Biol Chem. 1998;273(46):30208–17.

    Article  CAS  PubMed  Google Scholar 

  51. Balboa MA, Balsinde J, Johnson CA, Dennis EA. Regulation of arachidonic acid mobilization in lipopolysaccharide-activated P388D(1) macrophages by adenosine triphosphate. J Biol Chem. 1999;274(51):36764–8.

    Article  CAS  PubMed  Google Scholar 

  52. Barbera-Cremades M, Baroja-Mazo A, Gomez AI, Machado F, Di VF, Pelegrin P. P2X7 receptor-stimulation causes fever via PGE2 and IL-1beta release. FASEB J. 2012;26(7):2951–62.

    Article  CAS  PubMed  Google Scholar 

  53. Panupinthu N, Rogers JT, Zhao L, Solano-Flores LP, Possmayer F, Sims SM, et al. P2X7 receptors on osteoblasts couple to production of lysophosphatidic acid: a signaling axis promoting osteogenesis. J Cell Biol. 2008;181(5):859–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Costa-Junior HM, Marques-da-Silva C, Vieira FS, Moncao-Ribeiro LC, Coutinho-Silva R. Lipid metabolism modulation by the P2X7 receptor in the immune system and during the course of infection: new insights into the old view. Purinergic Signal. 2011;7(4):381–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Peters-Golden M, Brock TG. 5-lipoxygenase and FLAP. Prostaglandins Leukot Essent Fatty Acids. 2003;69(2-3):99–109.

    Article  CAS  PubMed  Google Scholar 

  56. Ago H, Kanaoka Y, Irikura D, Lam BK, Shimamura T, Austen KF, et al. Crystal structure of a human membrane protein involved in cysteinyl leukotriene biosynthesis. Nature. 2007;448(7153):609–12.

    Article  CAS  PubMed  Google Scholar 

  57. Lam BK, Gagnon L, Austen KF, Soberman RJ. The mechanism of leukotriene B4 export from human polymorphonuclear leukocytes. J Biol Chem. 1990;265(23):13438–41.

    CAS  PubMed  Google Scholar 

  58. Leier I, Jedlitschky G, Buchholz U, Cole SP, Deeley RG, Keppler D. The MRP gene encodes an ATP-dependent export pump for leukotriene C4 and structurally related conjugates. J Biol Chem. 1994;269(45):27807–10.

    CAS  PubMed  Google Scholar 

  59. Robbiani DF, Finch RA, Jager D, Muller WA, Sartorelli AC, Randolph GJ. The leukotriene C(4) transporter MRP1 regulates CCL19 (MIP-3beta, ELC)-dependent mobilization of dendritic cells to lymph nodes. Cell. 2000;103(5):757–68.

    Article  CAS  PubMed  Google Scholar 

  60. Murphy RC, Gijon MA. Biosynthesis and metabolism of leukotrienes. Biochem J. 2007;405(3):379–95.

    Article  CAS  PubMed  Google Scholar 

  61. Peters-Golden M, Canetti C, Mancuso P, Coffey MJ. Leukotrienes: underappreciated mediators of innate immune responses. J Immunol. 2005;174(2):589–94.

    Article  CAS  PubMed  Google Scholar 

  62. Ricciotti E, FitzGerald GA. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol. 2011;31(5):986–1000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Smith T, McCracken J, Shin YK, DeWitt D. Arachidonic acid and nonsteroidal anti-inflammatory drugs induce conformational changes in the human prostaglandin endoperoxide H2 synthase-2 (cyclooxygenase-2). J Biol Chem. 2000;275(51):40407–15.

    Article  CAS  PubMed  Google Scholar 

  64. Chandrasekharan NV, Dai H, Roos KL, Evanson NK, Tomsik J, Elton TS, et al. COX-3, a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs: cloning, structure, and expression. Proc Natl Acad Sci U S A. 2002;99(21):13926–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Dubois RN, Abramson SB, Crofford L, Gupta RA, Simon LS, Van De Putte LB, et al. Cyclooxygenase in biology and disease. FASEB J. 1998;12(12):1063–73.

    CAS  PubMed  Google Scholar 

  66. Scher JU, Pillinger MH. The anti-inflammatory effects of prostaglandins. J Investig Med. 2009;57(6):703–8.

    Article  CAS  PubMed  Google Scholar 

  67. Mukbel RM, Patten Jr C, Gibson K, Ghosh M, Petersen C, Jones DE. Macrophage killing of Leishmania amazonensis amastigotes requires both nitric oxide and superoxide. Am J Trop Med Hyg. 2007;76(4):669–75.

    CAS  PubMed  Google Scholar 

  68. Miralles GD, Stoeckle MY, McDermott DF, Finkelman FD, Murray HW. Th1 and Th2 cell-associated cytokines in experimental visceral leishmaniasis. Infect Immun. 1994;62(3):1058–63.

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Dey R, Majumder N, Bhattacharyya MS, Bhattacharjee S, Banerjee S, Roy S, et al. Induction of host protective Th1 immune response by chemokines in Leishmania donovani-infected BALB/c mice. Scand J Immunol. 2007;66(6):671–83.

    Article  CAS  PubMed  Google Scholar 

  70. Castellano LR, Filho DC, Argiro L, Dessein H, Prata A, Dessein A, et al. Th1/Th2 immune responses are associated with active cutaneous leishmaniasis and clinical cure is associated with strong interferon-gamma production. Hum Immunol. 2009;70(6):383–90.

    Article  CAS  PubMed  Google Scholar 

  71. Dudeck A, Suender CA, Kostka SL, von SE, Maurer M. Mast cells promote Th1 and Th17 responses by modulating dendritic cell maturation and function. Eur J Immunol. 2011;41(7):1883–93.

    Article  CAS  PubMed  Google Scholar 

  72. Katara GK, Ansari NA, Singh A, Ramesh V, Salotra P. Evidence for involvement of Th17 type responses in post kala azar dermal leishmaniasis (PKDL). PLoS Negl Trop Dis. 2012;6(6):e1703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Coutinho-Silva R, Ojcius DM. Role of extracellular nucleotides in the immune response against intracellular bacteria and protozoan parasites. Microbes Infect. 2012;14(14):1271–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Marques-da-Silva C, Chaves MM, Chaves SP, Figliuolo VR, Meyer-Fernandes JR, Corte-Real S, et al. Infection with Leishmania amazonensis upregulates purinergic receptor expression and induces host-cell susceptibility to UTP-mediated apoptosis. Cell Microbiol. 2011;13(9):1410–28.

    Article  CAS  PubMed  Google Scholar 

  75. Chaves SP, Torres-Santos EC, Marques C, Figliuolo VR, Persechini PM, Coutinho-Silva R, et al. Modulation of P2X(7) purinergic receptor in macrophages by Leishmania amazonensis and its role in parasite elimination. Microbes Infect. 2009;11(10-11):842–9.

    Article  CAS  PubMed  Google Scholar 

  76. Serezani CH, Perrela JH, Russo M, Peters-Golden M, Jancar S. Leukotrienes are essential for the control of Leishmania amazonensis infection and contribute to strain variation in susceptibility. J Immunol. 2006;177(5):3201–8.

    Article  CAS  PubMed  Google Scholar 

  77. Morato CI, da Silva IAJ, Borges AF, Dorta ML, Oliveira MA, Jancar S, et al. Essential role of leukotriene B4 on Leishmania (Viannia) braziliensis killing by human macrophages. Microbes Infect. 2014;16(11):945–53.

    Article  CAS  PubMed  Google Scholar 

  78. Tavares N, Afonso L, Suarez M, Ampuero M, Prates DB, Araujo-Santos T, et al. Degranulating neutrophils promote leukotriene B4 production by infected macrophages to kill Leishmania amazonensis parasites. J Immunol. 2016;196(4):1865–73.

    Article  CAS  PubMed  Google Scholar 

  79. Vishwakarma P, Parmar N, Yadav PK, Chandrakar P, Kar S. 15d-Prostaglandin J2 induced reactive oxygen species-mediated apoptosis during experimental visceral leishmaniasis. J Mol Med (Berl). 2016;94(6):695–710.

    Article  CAS  Google Scholar 

  80. Hu Y, Fisette PL, Denlinger LC, Guadarrama AG, Sommer JA, Proctor RA, et al. Purinergic receptor modulation of lipopolysaccharide signaling and inducible nitric-oxide synthase expression in RAW 264.7 macrophages. J Biol Chem. 1998;273(42):27170–5.

    Article  CAS  PubMed  Google Scholar 

  81. Martel-Gallegos G, Casas-Pruneda G, Ortega-Ortega F, Sanchez-Armass S, Olivares-Reyes JA, Diebold B, et al. Oxidative stress induced by P2X7 receptor stimulation in murine macrophages is mediated by c-Src/Pyk2 and ERK1/2. Biochim Biophys Acta. 2013;1830(10):4650–9.

    Article  CAS  PubMed  Google Scholar 

  82. Steiner DR, Gonzalez NC, Wood JG. Leukotriene B(4) promotes reactive oxidant generation and leukocyte adherence during acute hypoxia. J Appl Physiol (1985 ). 2001;91(3):1160–7.

    CAS  Google Scholar 

  83. Woo CH, You HJ, Cho SH, Eom YW, Chun JS, Yoo YJ, et al. Leukotriene B(4) stimulates Rac-ERK cascade to generate reactive oxygen species that mediates chemotaxis. J Biol Chem. 2002;277(10):8572–8.

    Article  CAS  PubMed  Google Scholar 

  84. Luchtefeld M, Drexler H, Schieffer B. 5-Lipoxygenase is involved in the angiotensin II-induced NAD(P)H-oxidase activation. Biochem Biophys Res Commun. 2003;308(3):668–72.

    Article  CAS  PubMed  Google Scholar 

  85. Lee JW, Kim JH. Activation of the leukotriene B4 receptor 2-reactive oxygen species (BLT2-ROS) cascade following detachment confers anoikis resistance in prostate cancer cells. J Biol Chem. 2013;288(42):30054–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Lee W, Su KH, Lee GR. Leukotrienes induce the migration of Th17 cells. Immunol Cell Biol. 2015;93(5):472–9.

    Article  CAS  PubMed  Google Scholar 

  87. Sacramento LA, Cunha FQ, de Almeida RP, da Silva JS, Carregaro V. Protective role of 5-lipoxigenase during Leishmania infantum infection is associated with Th17 subset. Biomed Res Int. 2014;2014:264270.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Toda A, Terawaki K, Yamazaki S, Saeki K, Shimizu T, Yokomizo T. Attenuated Th1 induction by dendritic cells from mice deficient in the leukotriene B4 receptor 1. Biochimie. 2010;92(6):682–91.

    Article  CAS  PubMed  Google Scholar 

  89. Sullivan JA, Jankowska-Gan E, Shi L, Roenneburg D, Hegde S, Greenspan DS, et al. Differential requirement for P2X7R function in IL-17 dependent vs. IL-17 independent cellular immune responses. Am J Transplant. 2014;14(7):1512–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Killeen ME, Ferris L, Kupetsky EA, Falo Jr L, Mathers AR. Signaling through purinergic receptors for ATP induces human cutaneous innate and adaptive Th17 responses: implications in the pathogenesis of psoriasis. J Immunol. 2013;190(8):4324–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Lonardoni MV, Barbieri CL, Russo M, Jancar S. Modulation of Leishmania (L.) amazonensis growth in cultured mouse macrophages by prostaglandins and platelet activating factor. Mediators Inflamm. 1994;3(2):137–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Barreto-de-Souza V, Pacheco GJ, Silva AR, Castro-Faria-Neto HC, Bozza PT, Saraiva EM, et al. Increased Leishmania replication in HIV-1-infected macrophages is mediated by tat protein through cyclooxygenase-2 expression and prostaglandin E2 synthesis. J Infect Dis. 2006;194(6):846–54.

    Article  CAS  PubMed  Google Scholar 

  93. Guimaraes ET, Santos LA, dos Ribeiro SR, Teixeira MM, dos Santos WL, Soares MB. Role of interleukin-4 and prostaglandin E2 in Leishmania amazonensis infection of BALB/c mice. Microbes Infect. 2006;8(5):1219–26.

    Article  CAS  PubMed  Google Scholar 

  94. Franca-Costa J, Van WJ, Boaventura VS, Luz NF, Malta-Santos H, Oliveira MC, et al. Arginase I, polyamine, and prostaglandin E2 pathways suppress the inflammatory response and contribute to diffuse cutaneous leishmaniasis. J Infect Dis. 2015;211(3):426–35.

    Article  PubMed  Google Scholar 

  95. Kabututu Z, Martin SK, Nozaki T, Kawazu S, Okada T, Munday CJ, et al. Prostaglandin production from arachidonic acid and evidence for a 9,11-endoperoxide prostaglandin H2 reductase in Leishmania. Int J Parasitol. 2003;33(2):221–8.

    Article  PubMed  Google Scholar 

  96. Araujo-Santos T, Rodriguez NE, Moura-Pontes S, Dixt UG, Abanades DR, Bozza PT, et al. Role of prostaglandin F2alpha production in lipid bodies from Leishmania infantum chagasi: insights on virulence. J Infect Dis. 2014;210(12):1951–61.

    Article  PubMed  Google Scholar 

  97. Griffon B, Cillard J, Chevanne M, Morel I, Cillard P, Sergent O. Macrophage-induced inhibition of nitric oxide production in primary rat hepatocyte cultures via prostaglandin E2 release. Hepatology. 1998;28(5):1300–8.

    Article  CAS  PubMed  Google Scholar 

  98. Betz M, Fox BS. Prostaglandin E2 inhibits production of Th1 lymphokines but not of Th2 lymphokines. J Immunol. 1991;146(1):108–13.

    CAS  PubMed  Google Scholar 

  99. Snijdewint FG, Kalinski P, Wierenga EA, Bos JD, Kapsenberg ML. Prostaglandin E2 differentially modulates cytokine secretion profiles of human T helper lymphocytes. J Immunol. 1993;150(12):5321–9.

    CAS  PubMed  Google Scholar 

  100. Hilkens CM, Vermeulen H, van Neerven RJ, Snijdewint FG, Wierenga EA, Kapsenberg ML. Differential modulation of T helper type 1 (Th1) and T helper type 2 (Th2) cytokine secretion by prostaglandin E2 critically depends on interleukin-2. Eur J Immunol. 1995;25(1):59–63.

    Article  CAS  PubMed  Google Scholar 

  101. Saha A, Biswas A, Srivastav S, Mukherjee M, Das PK, Ukil A. Prostaglandin E2 negatively regulates the production of inflammatory cytokines/chemokines and IL-17 in visceral leishmaniasis. J Immunol. 2014;193(5):2330–9.

    Article  CAS  PubMed  Google Scholar 

  102. Peng Z, Fernandez P, Wilder T, Yee H, Chiriboga L, Chan ES, et al. Ecto-5′-nucleotidase (CD73) -mediated extracellular adenosine production plays a critical role in hepatic fibrosis. FASEB J. 2008;22(7):2263–72.

    Article  CAS  PubMed  Google Scholar 

  103. Paletta-Silva R, Vieira DP, Vieira-Bernardo R, Majerowicz D, Gondim KC, Vannier-Santos MA, et al. Leishmania amazonensis: characterization of an ecto-3′-nucleotidase activity and its possible role in virulence. Exp Parasitol. 2011;129(3):277–83.

    Article  CAS  PubMed  Google Scholar 

  104. Rai AK, Thakur CP, Velpandian T, Sharma SK, Ghosh B, Mitra DK. High concentration of adenosine in human visceral leishmaniasis despite increased ADA and decreased CD73. Parasite Immunol. 2011;33(11):632–6.

    Article  CAS  PubMed  Google Scholar 

  105. Vieira DP, Paletta-Silva R, Saraiva EM, Lopes AH, Meyer-Fernandes JR. Leishmania chagasi: an ecto-3′-nucleotidase activity modulated by inorganic phosphate and its possible involvement in parasite-macrophage interaction. Exp Parasitol. 2011;127(3):702–7.

    Article  CAS  PubMed  Google Scholar 

  106. Leite PM, Gomes RS, Figueiredo AB, Serafim TD, Tafuri WL, de Souza CC, et al. Ecto-nucleotidase activities of promastigotes from Leishmania (Viannia) braziliensis relates to parasite infectivity and disease clinical outcome. PLoS Negl Trop Dis. 2012;6(10), e1850.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Pinheiro CM, Martins-Duarte ES, Ferraro RB, Fonseca de Souza AL, Gomes MT, Lopes AH, et al. Leishmania amazonensis: Biological and biochemical characterization of ecto-nucleoside triphosphate diphosphohydrolase activities. Exp Parasitol. 2006;114(1):16–25.

    Article  CAS  PubMed  Google Scholar 

  108. Giarola NL, Silveira TS, Inacio JD, Vieira LP, Almeida-Amaral EE, Meyer-Fernandes JR. Leishmania amazonensis: Increase in ecto-ATPase activity and parasite burden of vinblastine-resistant protozoa. Exp Parasitol.2014;146:25–33.

    Article  CAS  PubMed  Google Scholar 

  109. Gomes RS, de Carvalho LC, de S V, Fietto JL, Afonso LC. E-NTPDase (ecto-nucleoside triphosphate diphosphohydrolase) of Leishmania amazonensis inhibits macrophage activation. Microbes Infect. 2015;17(4):295–303.

    Article  CAS  PubMed  Google Scholar 

  110. Berredo-Pinho M, Peres-Sampaio CE, Chrispim PP, Belmont-Firpo R, Lemos AP, Martiny A, et al. A Mg-dependent ecto-ATPase in Leishmania amazonensis and its possible role in adenosine acquisition and virulence. Arch Biochem Biophys. 2001;391(1):16–24.

    Article  CAS  PubMed  Google Scholar 

  111. Figueiredo AB, Serafim TD, Marques-da-Silva EA, Meyer-Fernandes JR, Afonso LC. Leishmania amazonensis impairs DC function by inhibiting CD40 expression via A2B adenosine receptor activation. Eur J Immunol. 2012;42(5):1203–15.

    Article  CAS  PubMed  Google Scholar 

  112. Cadieux JS, Leclerc P, St-Onge M, Dussault AA, Laflamme C, Picard S, et al. Potentiation of neutrophil cyclooxygenase-2 by adenosine: an early anti-inflammatory signal. J Cell Sci. 2005;118(Pt 7):1437–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Pouliot M, Fiset ME, Masse M, Naccache PH, Borgeat P. Adenosine up-regulates cyclooxygenase-2 in human granulocytes: impact on the balance of eicosanoid generation. J Immunol. 2002;169(9):5279–86.

    Article  PubMed  Google Scholar 

  114. MacKenzie KF, Clark K, Naqvi S, McGuire VA, Noehren G, Kristariyanto Y, et al. PGE(2) induces macrophage IL-10 production and a regulatory-like phenotype via a protein kinase A-SIK-CRTC3 pathway. J Immunol. 2013;190(2):565–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Nemeth ZH, Lutz CS, Csoka B, Deitch EA, Leibovich SJ, Gause WC, et al. Adenosine augments IL-10 production by macrophages through an A2B receptor-mediated posttranscriptional mechanism. J Immunol. 2005;175(12):8260–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Hasko G, Kuhel DG, Chen JF, Schwarzschild MA, Deitch EA, Mabley JG, et al. Adenosine inhibits IL-12 and TNF-[alpha] production via adenosine A2a receptor-dependent and independent mechanisms. FASEB J. 2000;14(13):2065–74.

    Article  CAS  PubMed  Google Scholar 

  117. Vassiliou E, Jing H, Ganea D. Prostaglandin E2 inhibits TNF production in murine bone marrow-derived dendritic cells. Cell Immunol. 2003;223(2):120–32.

    Article  CAS  PubMed  Google Scholar 

  118. Flamand N, Boudreault S, Picard S, Austin M, Surette ME, Plante H, et al. Adenosine, a potent natural suppressor of arachidonic acid release and leukotriene biosynthesis in human neutrophils. Am J Respir Crit Care Med. 2000;161(2 Pt 2):S88–94.

    Article  CAS  PubMed  Google Scholar 

  119. Krump E, Picard S, Mancini J, Borgeat P. Suppression of leukotriene B4 biosynthesis by endogenous adenosine in ligand-activated human neutrophils. J Exp Med. 1997;186(8):1401–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Surette ME, Krump E, Picard S, Borgeat P. Activation of leukotriene synthesis in human neutrophils by exogenous arachidonic acid: inhibition by adenosine A(2a) receptor agonists and crucial role of autocrine activation by leukotriene B(4). Mol Pharmacol. 1999;56(5):1055–62.

    CAS  PubMed  Google Scholar 

  121. Krump E, Lemay G, Borgeat P. Adenosine A2 receptor-induced inhibition of leukotriene B4 synthesis in whole blood ex vivo. Br J Pharmacol. 1996;117(8):1639–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Peters NC, Sacks DL. The impact of vector-mediated neutrophil recruitment on cutaneous leishmaniasis. Cell Microbiol. 2009;11(9):1290–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Laufs H, Muller K, Fleischer J, Reiling N, Jahnke N, Jensenius JC, et al. Intracellular survival of Leishmania major in neutrophil granulocytes after uptake in the absence of heat-labile serum factors. Infect Immun. 2002;70(2):826–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Ribeiro-Gomes FL, Peters NC, Debrabant A, Sacks DL. Efficient capture of infected neutrophils by dendritic cells in the skin inhibits the early anti-Leishmania response. PLoS Pathog. 2012;8(2):e1002536.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Novais FO, Santiago RC, Bafica A, Khouri R, Afonso L, Borges VM, et al. Neutrophils and macrophages cooperate in host resistance against Leishmania braziliensis infection. J Immunol. 2009;183(12):8088–98.

    Article  CAS  PubMed  Google Scholar 

  126. Mahamed DA, Mills JH, Egan CE, Denkers EY, Bynoe MS. CD73-generated adenosine facilitates Toxoplasma gondii differentiation to long-lived tissue cysts in the central nervous system. Proc Natl Acad Sci U S A. 2012;109(40):16312–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Thammavongsa V, Kern JW, Missiakas DM, Schneewind O. Staphylococcus aureus synthesizes adenosine to escape host immune responses. J Exp Med. 2009;206(11):2417–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Firon A, Dinis M, Raynal B, Poyart C, Trieu-Cuot P, Kaminski PA. Extracellular nucleotide catabolism by the Group B Streptococcus ectonucleotidase NudP increases bacterial survival in blood. J Biol Chem. 2014;289(9):5479–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Belkaid Y, Kamhawi S, Modi G, Valenzuela J, Noben-Trauth N, Rowton E, et al. Development of a natural model of cutaneous leishmaniasis: powerful effects of vector saliva and saliva preexposure on the long-term outcome of Leishmania major infection in the mouse ear dermis. J Exp Med. 1998;188(10):1941–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Kamhawi S. The biological and immunomodulatory properties of sand fly saliva and its role in the establishment of Leishmania infections. Microbes Infect. 2000;2(14):1765–73.

    Article  CAS  PubMed  Google Scholar 

  131. Hall LR, Titus RG. Sand fly vector saliva selectively modulates macrophage functions that inhibit killing of Leishmania major and nitric oxide production. J Immunol. 1995;155(7):3501–6.

    CAS  PubMed  Google Scholar 

  132. Prates DB, Araujo-Santos T, Luz NF, Andrade BB, Franca-Costa J, Afonso L, et al. Lutzomyia longipalpis saliva drives apoptosis and enhances parasite burden in neutrophils. J Leukoc Biol. 2011;90(3):575–82.

    Article  CAS  PubMed  Google Scholar 

  133. Charlab R, Valenzuela JG, Rowton ED, Ribeiro JM. Toward an understanding of the biochemical and pharmacological complexity of the saliva of a hematophagous sand fly Lutzomyia longipalpis. Proc Natl Acad Sci USA. 1999;96(26):15155–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. de Moura TR, Oliveira F, Rodrigues GC, Carneiro MW, Fukutani KF, Novais FO, et al. Immunity to Lutzomyia intermedia saliva modulates the inflammatory environment induced by Leishmania braziliensis. PLoS Negl Trop Dis. 2010;4(6):e712.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Katz O, Waitumbi JN, Zer R, Warburg A. Adenosine, AMP, and protein phosphatase activity in sandfly saliva. Am J Trop Med Hyg. 2000;62(1):145–50.

    CAS  PubMed  Google Scholar 

  136. Carregaro V, Ribeiro JM, Valenzuela JG, Souza-Junior DL, Costa DL, Oliveira CJ, et al. Nucleosides present on phlebotomine saliva induce immunossuppression and promote the infection establishment. PLoS Negl Trop Dis. 2015;9(4):e0003600.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Atayde VD, Aslan H, Townsend S, Hassani K, Kamhawi S, Olivier M. Exosome secretion by the parasitic protozoan leishmania within the sand fly midgut. Cell Rep. 2015;13(5):957–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Araujo-Santos T, Prates DB, Andrade BB, Nascimento DO, Clarencio J, Entringer PF, et al. Lutzomyia longipalpis saliva triggers lipid body formation and prostaglandin E(2) production in murine macrophages. PLoS Negl Trop Dis. 2010;4(11):e873.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  139. Araujo-Santos T, Prates DB, Franca-Costa J, Luz NF, Andrade BB, Miranda JC, et al. Prostaglandin E2/leukotriene B4 balance induced by Lutzomyia longipalpis saliva favors Leishmania infantum infection. Parasit Vectors. 2014;7:601.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

We thank Ludmilla Dellatorre for help in preparing the figures, Dr Larry Johnson for revision of English language and CNPq, CAPES and FAPERJ for financial support.

Funding

This work was supported by the Conselho Nacional de Deselvimento Científico e Tecnológico (CNPq), Coordenação de Aperfeiçoamento de Nível Superior (CAPES) and Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ) (grant number E-26/111.182/2011).

Availability of data and material

Not applicable.

Authors’ contributions

MMC carried out the literature search, prepared the draft of the manuscript and wrote the paper. CC contributed to writing the paper and critically revised the manuscript. RCS contributed to writing the paper and critically revised the manuscript. All authors read and approved the final manuscript.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Disclosures

The authors have no financial conflicts of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Robson Coutinho-Silva.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chaves, M.M., Canetti, C. & Coutinho-Silva, R. Crosstalk between purinergic receptors and lipid mediators in leishmaniasis. Parasites Vectors 9, 489 (2016). https://doi.org/10.1186/s13071-016-1781-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13071-016-1781-1

Keywords