Abstract
Diseases caused by Leishmania present a worldwide problem, and current therapeutic approaches are unable to achieve a sterile cure. Leishmania is able to persist in host cells by evading or exploiting host immune mechanisms. A thorough understanding of these mechanisms could lead to better strategies for effective management of Leishmania infections. Current research has focused on parasite modification of host cell signaling pathways, entry into phagocytic cells, and modulation of cytokine and chemokine profiles that alter immune cell activation and trafficking to sites of infection. Immuno-therapeutic approaches that target these mechanisms of immune evasion by Leishmania offer promising areas for preclinical and clinical research.
Diseases caused by Leishmania are a major global health problem as over 12 million people currently suffer from leishmaniasis, with an incidence rate of approximately 2 million annually, according to recent estimates (www.who.int/tdr). It is transmitted by sandflies and presents a wide range of clinical manifestations that depend on the specie and strain of Leishmania, degree of virulence, and the immunological state of the host. Cutaneous leishmaniasis (CL) manifests as localized skin lesions that may resolve but can become chronic, leading to severe tissue destruction and disfigurement. Lesions could disseminate in immunocompromised patients giving rise to the diffuse cutaneous leishmaniasis (DCL) (Desjeux, 2004). This disease is caused by Leishmania major (the Middle East and Mediterranean Region), Leishmania mexicana (Central America), and Leishmania amazonensis (South America). Leishmania tropica and Leishmania aethiopica also cause CL in the Old World. Mucocutaneous leishmaniasis caused by Leishmania braziliensis is endemic in South America and is clinically characterized by the involvement of the nasal and oropharyngeal mucosa with extensive tissue destruction due to inflammation. Visceral leishmaniasis (VL), the most severe form of leishmaniasis, is caused by Leishmania donovani and Leishmania chagasi in the Old and New worlds, respectively. Clinical manifestations include hepatomegaly and splenomegaly due to parasite infiltration of the liver and spleen and if left untreated, it is almost always fatal (www.who.int/tdr) (Alexander, Satoskar, & Russell, 1999; Awasthi, Mathur, & Saha, 2004; Desjeux, 2004).
Chemotherapeutic and prophylactic approaches to the management of the various forms of leishmaniasis have been problematic due to the tendency of the parasite to persist within host cells and subsequent failure to achieve a sterile cure. Such latent or chronic forms of the disease present obvious dangers when the hosts’ immune system is compromised. The key to effective management of Leishmania infections therefore depends on a thorough understanding of the immunological basis for parasite persistence within host cells. Interestingly, macrophages which are the primary immune cells involved in the eradication of Leishmania in a mammalian host are the targets for the parasite. How Leishmania is able to survive and thrive in this hostile environment as well as capitalize on host defense mechanisms to favor the establishment of disease will be addressed in this review.
1. IMMUNITY TO LEISHMANIA
Successful elimination of Leishmania depends on the coordinated action of various players of the immune system. From promastigote entry into the blood stream after a sandfly bite to its final mammalian cellular target as an amastigote, the battle between disease establishment and parasite eradication will partly be decided by the ability of Leishmania to evade host immunity. Components of host defense important for the eradication of Leishmania include both elements of the innate and adaptive immune system.
1.1. The Complement System
It has long been known that the complement system plays a significant role in the eradication of promastigotes in the blood stream of an infected host (Rezai, Sher, & Gettner, 1969). Recent studies have shown that susceptibility of Leishmania parasites to complement-mediated lysis in vitro is directly related to the concentration of serum complement (Moreno et al., 2007). Although the classical pathway of the complement system is activated by Leishmania, complement-mediated destruction of parasites is generally amplified by the alternate pathway (Hoover, Berger, Nacy, Hockmeyer, & Meltzer, 1984). Amastigotes from L. donovani are more resistant than L. tropica amastigotes to lysis by complement, suggesting that some Leishmania spp. might actively resist this process (Hoover et al., 1984). It should be noted that while unfractionated promastigotes are highly susceptible to lysis by complement, infective metacyclic promastigotes are generally more resistant (Puentes, Sacks, da Silva, & Joiner, 1988).
1.2. Cells of Innate Immunity
Neutrophils are the first host cells to reach the site of Leishmania infection within a few hours of inoculation by a sandfly bite (Müller et al., 2001; Pompeu, Freitas, Santos, Khouri, & Barral-Netto, 1991). They have been shown to actively engulf Leishmania promastigotes (Mollinedo, Janssen, de la Iglesia-Vicente, Villa-Pulgarin, & Calafat, 2010; Pearson & Steigbigel, 1981) and produce an array of microbicidal factors against Leishmania such as nitric oxide (Charmoy et al., 2007), neutrophil elastase (NE) (Ribeiro-Gomes et al., 2007), platelet activating factor (Camussi, Bussolino, Salvidio, & Baglioni, 1987), and neutrophil extracellular traps (Guimarães-Costa et al., 2009). Neutrophils generally have a protective role in most forms of Leishmania infections (de Souza Carmo, Katz, & Barbiéri, 2010; Novais et al., 2009), although outcomes are dependent on the Leishmania strain, the genetic background of the host and the apoptotic or necrotic state of the neutrophils (Afonso et al., 2008; de Souza Carmo et al., 2010; Filardy et al., 2010; Novais et al., 2009; Ribeiro-Gomes et al., 2004). In vivo depletion of neutrophils during L. major infection results in an increase in parasite load in resistant mice (C57BL/6 and C3H/HeJ mice), but susceptible mice (BALB/c mice) show a reduction in parasite load (Ribeiro-Gomes et al., 2004; Tacchini-Cottier et al., 2000).
Early on during Leishmania infection, natural killer (NK) cells are also recruited to the infected site after neutrophil recruitment (Müller et al., 2001; Thalhofer, Chen, Sudan, Love-Homan, & Wilson, 2011). They are the primary source of early IFN-γ that favors the Th1 differentiation of CD4+ T cells (Scharton & Scott, 1993) and restricts early parasite dissemination (Diefenbach et al., 1998; Laskay, Diefenbach, Röllinghoff, & Solbach, 1995). NK cells can also mediate direct parasite lysis through its cytotoxic activity (Lieke et al., 2011) and subsequently contribute to cytokine-mediated inducible nitric oxide synthase (iNOS) induction in Leishmania-infected macrophages (Prajeeth, Haeberlein, Sebald, Schleicher, & Bogdan, 2011). Studies using NK cell deficient mice have shown the importance of these cells in the containment of L. donovani infection but they are dispensable in models of cutaneous leishmaniasis (Kirkpatrick & Farrell, 1982; Satoskar et al., 1999). In humans, NK cell population has been shown to decrease in cases of progressive disease (Pereira et al., 2009; Peruhype-Magalhães et al., 2005), while sites of healing lesions show an infiltration of CD56+ NK cells (Pereira et al., 2009), which suggests a protective role for NK cell population in human leishmaniasis.
Natural killer T (NKT) cells, a specialized subset of T lymphocytes involved in innate immunity to pathogens, also play important roles in the immune response during the early stages of Leishmania infection (Amprey et al., 2004; Ishikawa et al., 2000), although these roles vary depending on the specie of Leishmania and strain of mice. Leishmania surface glyco-conjugates and glycoinositol are recognized by CD1d restricted NKT cells (Amprey et al., 2004; Mattner, Donhauser, Werner-Felmayer, & Bogdan, 2006) which contribute to hepatic clearance of L. donovani in BALB/c mice through the induction of IFN-γ (Amprey et al., 2004), but exacerbates the disease in C57BL/6 mice (Stanley et al., 2008). In L. major infection, protection by NKT cells seems to be organ specific, where they contribute to parasite control in skin lesions and the spleen but not in the lymph nodes (Mattner et al., 2006). In humans, an increase in iNKT cell frequency in the bone marrow of VL patients is observed as the disease progresses, which decreases following treatment (Rai, Thakur, Seth, & Mitra, 2011).
1.3. Cells of Adaptive Immunity
Lymphocytes are involved in adaptive immune responses to Leishmania infection, primarily through the elaboration of cytokines that activate or dampen the antiparasitic activity of macrophages. It is well known that T cells play a major role in immunity to the various forms of Leishmania. Generally, IFN-γ-producing Th1 cells are essential to the resolution of infection with L. major, where they induce nitric oxide production in macrophages. On the other hand, susceptibility is associated with the production of cytokines produced by Th2 cells such as IL-4 and IL-13. Indeed, genetically resistant (such as C57BL/6) or susceptible (such as BALB/c) mice to L. major infection are characterized by their ability to produce Th1 or Th2 cytokine profiles respectively. This is also the case in visceral leishmaniasis caused by L. donovani, in mice and humans where IFN-γ-producing Th1 cells protect against severe disease (Kushawaha, Gupta, Sundar, Sahasrabuddhe, & Dube, 2011). In human cutaneous leishmaniasis IFN-γ production by CD8+ T cells seems to contribute to disease resolution (Mary, Auriault, Faugère, & Dessein, 1999; Nateghi Rostami et al., 2010). However, as demonstrated by Leishmania infection models using CD4 or CD8 deficient mice, while CD4 T cells are required for resolution of disease (Chakkalath et al., 1995), the role of CD8 T cells in immunity to cutaneous or visceral leishmaniasis seems to depend on the model used (Belkaid, Von Stebut, et al., 2002; Erb, Blank, Ritter, Bluethmann, & Moll, 1996; Gomes-Pereira, Rodrigues, Rolão, Almeida, & Santos-Gomes, 2004; Huber, Timms, Mak, Röllinghoff, & Lohoff, 1998; Tsagozis, Karagouni, & Dotsika, 2005).
Studies using mouse models that affect the activity and migration of regulatory T cells show that they play key roles in regulating the immune response to L. major infection (Liu et al., 2009; Suffia, Reckling, Salay, & Belkaid, 2005; Yurchenko et al., 2006). On the one hand, they dampen the effector response of CD4+ T cells partly mediated through IL-10 expression by Tregs. This results in parasite persistence even in resistant C57BL/6 mice, which has implications in the case of disease reactivation. On the other hand, by allowing parasite persistence, Tregs contribute to the maintenance of long term immunity to L. major (Belkaid, 2003; Belkaid, Piccirillo, Mendez, Shevach, & Sacks, 2002). This balance between effector and regulatory T cells could potentially be exploited by Leishmania parasites to evade host immune responses.
Other immune cells are also involved in controlling immunity to Leishmania. B cells seem to play a detrimental role in the early stages of L. donovani infection, as shown by significantly reduced parasite burdens in B cell deficient mice (Smelt, Cotterell, Engwerda, & Kaye, 2000). Marginal zone B cells in particular have been shown to dampen the cytotoxic activity of antigen specific CD8 T cells as well as the frequency of IFN gamma producing CD4+ T cells (Bankoti, Gupta, Levchenko, & Stäger, 2012), thus contributing to increased parasite loads. Recent studies on myeloid-derived suppressors cells show that they contribute to resistance to L. major in an NO dependent manner, despite their ability to suppress T cell activation (Pereira et al., 2011). During L. major infection, dendritic cell subsets as well as their differentiation state play diverse roles in modulating the adaptive immune and affecting the outcome of the disease (Kautz-Neu et al., 2011; Wiethe et al., 2008).
1.4. Cytokines and Chemokines
As shown by a variety of cytokine and chemokine knockout mouse models, cytokines and chemokines play a huge role in immunity to a host of infectious diseases including Leishmania infections. While some of these secreted protein immune-modulators are involved in the activation and differentiation of immune cells important in parasite clearance (such as IL-12, TNF-α, IFN-γ) (Mattner et al., 1996; Swihart et al., 1995), others could either dampen the immune response against Leishmania or activate and differentiate immune cells that will ultimately favor the persistence of the parasite (such as IL-4, IL-10, IL-13, TGF-β) (Kopf et al., 1996). Cytokines are produced by immune or infected cells and exert their function by activating other cells to release molecules that inhibit or favor the growth of Leishmania. Chemokines and chemokine receptors are involved in trafficking of immune cells to inflammatory sites. Indeed, many researchers have suggested their use in the immuno-prophylaxis or therapy of leishmaniasis (Gupta, Majumdar, et al., 2011).
2. MECHANISMS OF IMMUNE EVASION
Like any successful pathogen, Leishmania has developed strategies to evade host immune mechanisms in order to survive within the host. A significant number of virulence factors discovered in Leishmania are directed against circumventing the host immune response. The ability of Leishmania to maintain a chronic infectious state within its host depends to a large extent on its immune evasion potential. Indeed, the ongoing battle between the robust immune response mounted by a host and the counter evasion strategies by the parasite will ultimately decide the fate of the disease. The mechanisms of immune evasion by Leishmania species include the following:
2.1. Modification of the Complement System and Phagocytosis
Once the female sandfly injects Leishmania promastigotes into the mammalian host, it becomes imperative for the parasite to escape or deactivate the host complement system before entering in the macrophages. Leishmania is able to evade the host’s complement system using a variety of mechanisms. Unlike noninfective procyclic promastigotes, infective metacyclic promastigotes prevent the insertion of the C5–C9 membrane attack complex making them highly resistant to complement-mediated lysis (Puentes, Da Silva, Sacks, Hammer, & Joiner, 1990). This parasite stage-specific difference in complement evasion is due to a developmental modification resulting in the elongation of lipophosphoglycan (LPG) in metacyclic Leishmania (Sacks, Pimenta, McConville, Schneider, & Turco, 1995). Among the various stages of Leishmania, metacyclic promastigotes show the highest expression of protein kinases, which phosphorylate complement proteins C3, C5, and C9, resulting in the deactivation of classical and alternative complement pathways (Hermoso, Fishelson, Becker, Hirschberg, & Jaffe, 1991). The activity of Leishmania glycoprotein 63 (GP63), a metalloproteinase found abundantly on the surface of metacyclic promastigotes, is pivotal for resisting complement lysis. GP63 cleaves the C3b to an inactive form, C3bi on the surface membrane of the parasite, thereby hindering the formation of C5 convertase (Brittingham et al., 1995).
C3bi also serves as an opsonin, facilitating the uptake of the parasite by binding to complement receptor 3 (CR3) and transiently to CR1 on the surface of macrophages (Kane & Mosser, 2000; Mosser & Edelson, 1985). Attachment via CR3 rather than CR1 is advantageous to the parasite as CR3 ligation even in the absence of Leishmania inhibits the production of IL-12 (Marth & Kelsall, 1997). Thus Leishmania is able to exploit CR3-mediated phagocytosis, facilitating a ‘silent entry’ into macrophages thereby evading the host’s protective immune response (Da Silva, Hall, Joiner, & Sacks, 1989; Wright & Silverstein, 1983). To further support the role of CR3 in mediating host susceptibility to Leishmania, a recent study using CR3 deficient BALB/c mice showed an increased resistance to L. major infection (Carter, Whitcomb, Campbell, Mukbel, & McDowell, 2009).
Although macrophages are involved in the eradication of Leishmania, they are the primary host cells targeted by the parasite for survival and proliferation. As such Leishmania possess a variety of cell surface molecules that enable them localize to macrophages. LPG on the surface of Leishmania promastigotes can facilitate binding to mannosyl/fucosyl receptors (Blackwell et al., 1985), complement reactive protein (Culley, Harris, Kaye, McAdam, & Raynes, 1996), and CR4 (Talamás-Rohana, Wright, Lennartz, & Russell, 1990) on macrophage membranes. Furthermore, Leishmania glycosylinositol phospholipid (GIPL) and GP63 have important roles in the attachment of parasites to macrophages (Brittingham et al., 1995; Suzuki, Tanaka, Toledo, Takahashi, & Straus, 2002). Leishmania amastigotes, unlike promastigotes, can be internalized using phosphatidyl serine and Fc receptors although additional receptors may also participate in the entry process (de Freitas Balanco et al., 2001; Guy & Belosevic, 1993; Weingartner et al., 2012). Recently, our group demonstrated that the phosphoinositide 3-kinase gamma (PI3-Kγ) signaling pathway is exploited by L. mexicana to facilitate parasite entry into macrophages and progression of disease. Further, therapy using the PI3-Kγ specific inhibitor AS-605240 protects against cutaneous leishmaniasis caused by L. mexicana (Cummings et al., 2012).
2.2. Alteration of Toll-Like Receptor Pathways
Toll-like receptors (TLRs) expressed on the cells of the innate immune system are critical for recognition of pathogen-associated molecular patterns. Initial interaction of the parasite with different TLR’s dictates the outcome of the infection (Faria, Reis, & Lima, 2012). TLR2 on host macrophages recognizes Leishmania promastigote-derived LPG (de Veer et al., 2003; Flandin, Chano, & Descoteaux, 2006) and amastigote-specific antigens (Srivastava et al., 2012). LPG interaction with TLR2 results in the induction of TNF-α (Flandin et al., 2006), IL-12 (Kavoosi, Ardes-tani, & Kariminia, 2009), NO (Kavoosi, Ardestani, Kariminia, & Alimo-hammadian, 2010), and reactive oxygen species (ROS) (Kavoosi et al., 2009). TLR2 agonists which activate this TLR2 signaling pathway has been utilized in inducing a host protective immune response resulting in parasite clearance from L. donovani infected macrophages (Bhattacharya et al., 2010).
To subvert this inflammatory response L. major recruits suppressors of the cytokine signaling family proteins, SOCS-1 and SOCS-3, which negatively regulates TLR2 induced cytokine induction (de Veer et al., 2003). Another mechanism of TLR2-mediated suppression by Leishmania involves the activation of host de-ubiquitinating enzyme A20 by L. donovani promastigotes resulting in the impairment of TLR2-mediated release of IL-12 and TNF-α as the parasite interferes with the ubiquitination of TRAF6 (Srivastav, Kar, Chande, Mukhopadhyaya, & Das, 2012). On the other hand, L. amazonensis capitalize on TLR2 signaling to facilitate the establishment of infection, by increasing the expression of double stranded RNA dependent protein kinase (PKR) and IFN-β. IFN-β increases superoxide dismutase 1 levels which inhibits superoxide-dependent parasite killing and augments parasite replication (Khouri et al., 2009; Vivarini et al., 2011). Furthermore, a similar study with TLR2-deficient mice showed an essential role of TLR2 in lesion development during L. braziliensis infection (Vargas-Inchaustegui et al., 2009). These findings suggest multiple mechanisms employed by different species of Leishmania to alter TLR2 signaling and enhance parasite establishment.
TLR4 plays a critical role in shaping the host immune response during Leishmania infection. Studies with TLR4−/− mice demonstrate the role of TLR4 in the control of L. major infection (Kropf, Freudenberg, Modolell, et al., 2004, Kropf, Freudenberg, Kalis, et al., 2004). Glyco-sphingophospholipid (GSPL) and proteoglycolipid complex (P8GLC) from Leishmania induce TLR4, promoting a strong antiparasitic immune response (Karmakar, Bhaumik, Paul, & De, 2012; Whitaker, Colmenares, Pestana, & McMahon-Pratt, 2008). There is a strong TNF-α response upon P8GLC-TLR4 engagement in macrophages infected with Leishmania pifanoi amastigotes and in vivo treatment with P8GLC showed enhanced parasite clearance in TLR4-competent infected mice compared to TLR4−/−-deficient infected mice (Whitaker et al., 2008). Similar results were obtained using GSPL treatment in L. donovani infection (Karmakar, Paul, & De, 2011).
Leishmania has devised mechanisms to alter TLR4 signaling pathways to favor establishment of infection. During L. donovani infection, TLR4-mediated macrophage activation is suppressed through the release of TGF-β that activates the ubiquitin editing enzyme A20 and Src homology 2 domain phosphotyrosine phosphatase 1 (SHP-1) (Das et al., 2012). Leishmania major utilizes its inhibitors of serine protease (ISP) to prevent NE-mediated TLR4 activation, inhibiting Leishmania uptake and killing by host macrophages (Faria et al., 2011; Ribeiro-Gomes et al., 2007). On the other hand, L. mexicana capitalizes on TLR4 signaling to inhibit the production of IL-12 by infected macrophages and promotes parasite establishment (Shweash et al., 2011).
Other TLRs involved in infection with Leishmania include TLR3 and TLR9. Endogenous TLR3 binds to double stranded RNA and has been shown to be activated during Leishmania infection. A virulent strain of Leishmania guyanensis and Leishmania vianna which harbor Leishmania RNA virus (LRV1) has been shown to activate the TLR3-TRIF–dependent pathway essential for increased pro-inflammatory mediator expression after macrophage infection (Hartley, Ronet, Zangger, Beverley, & Fasel, 2012). Interestingly, TLR3-mediated immune responses rendered mice more susceptible to infection showing increased footpad swelling and parasitemia along with increased metastasis (Ives et al., 2011).
2.3. Surviving in the Phagosome
To survive inside the macrophage, Leishmania must resist the harsh conditions created by the phagocyte, including acidic pH, elevated temperature, and increased oxidative/nitrosative stress. Upon phagocytosis, Leishmania promastigotes are internalized into endosomal compartments, where they transform into amastigotes. In order to escape the hostile environment of the phagolysosome, promastigotes transiently prevent fusion of the phagosome and lysosome thereby delaying or inhibiting endosomal maturation as observed by the late expression of Rab7 and LAMP-1 (Olivier, Gregory, & Forget, 2005; Scianimanico et al., 1999). LPG on Leishmania promastigotes inhibits endosome maturation by inducing periphagosomal F-actin accumulation (Holm, Tejle, Magnusson, Descoteaux, & Rasmusson, 2001). LPG also prevents acidification of the phagosome by interfering with the V-ATPase pump, which allows promastigotes to differentiate into resistant amastigotes (Vinet, Fukuda, Turco, & Descoteaux, 2009). Scavenging on host sphingolipids is another survival strategy employed by Leishmania amastigotes for counteracting the acidic environment of the phagolysosome (Ali, Harding, & Denny, 2012). In order to dilute the leishmanicidal effect of nitric oxide, Leishmania regulates the lysosomal trafficking (LYST) protein resulting in the formation of large parasitophorous vacuoles (Wilson et al., 2008).
Leishmania amastigotes require iron for metabolism and replication, therefore acquisition of iron is critical for its survival within the macrophage phagolysosome (Huynh & Andrews, 2008). In murine macrophages, Nramp1 functions as an efflux pump that translocates Fe2+ from the phagolysosome into the cytosol thereby restricting its availability to the parasite. Leishmania counteracts this effect by the activation of its own iron transporters, LIT1 and LIT2 which effectively compete with the host’s iron sequestering mechanism (Kaye & Scott, 2011). Arginine is also an essential growth factor required by intracellular Leishmania amastigotes for the synthesis of polyamines, but is also used by macrophages for microbicidal nitric oxide production (Iniesta, Gómez-Nieto, & Corraliza, 2001; Kropf, Herath, Weber, Modolell, & Müller, 2003). As demonstrated by studies using gene deficient mutants of Leishmania, the parasite encoded arginase enzyme not only facilitates growth by supplying essential nutrients to the parasite through the polyamine pathway, but also attenuates the iNOS-dependent killing mechanism of infected macrophages by competing for available intracellular arginine (Gaur et al., 2007; Reguera, Balaña-Fouce, Showalter, Hickerson, & Beverley, 2009). These virulence factors expressed by Leishmania amastigotes enable the parasite to evade the host’s cellular immune response and survive in the phagosome.
2.4. Defective Antigen Presentation and Co-stimulation
Virulent stages of Leishmania have the capacity to attenuate T cell-mediated immune responses by regulating the expression of leishmanial antigen loaded major histocompatibility complex (MHC) molecules on antigen presenting cells. They accomplish this by antigen sequestration or interference with the loading of antigens onto MHC class II molecules (Fruth, Solioz, & Louis, 1993; Kima, Soong, Chicharro, Ruddle, & McMahon-Pratt, 1996; Prina, Lang, Glaichenhaus, & Antoine, 1996). Studies on presentation of the Leishmania antigen, Leishmania homolog of receptors for activated C kinase (LACK), by infected macrophages showed a transient yet strong LACK-specific T cell response when infected with stationary or log phase Leishmania promastigotes. On the other hand, murine macrophages infected with either Leishmania metacyclic promastigotes or amastigotes showed a weak or absent LACK-specific T cell activation respectively (Courret et al., 1999). Membrane lipid rafts are important platforms for antigen presentation as it concentrates MHC class II molecules into microdomains, which allow efficient antigen presentation at low peptide densities. Leishmania donovani increases the fluidity of lipid rafts in macrophages resulting in defective antigen presentation and diminished T cells responses (Chakraborty et al., 2005). MHC class II, but not class I molecules have been shown to be important in host resistance against Leishmania (Huber et al., 1998; Locksley, Reiner, Hatam, Littman, & Killeen, 1993), and only MHC II molecules appear in the parasitophorous vacuole of an infected macrophage (Lang et al., 1994). MHC II molecules, located in specific organelles called megasomes, are endocytosed by Leishmania amastigotes and degraded by cysteine proteases (De Souza Leao, Lang, Prina, Hellio, & Antoine, 1995). These examples highlight the ability of Leishmania to evade recognition by T lymphocytes by interfering with macrophage antigen presentation.
Co-stimulatory molecules such as B7-1, B7-2, and CD40 expressed on macrophages are also critical for setting up antiparasitic T cell responses (Alexander et al., 1999; Bogdan, Gessner, Solbach, & Röllinghoff, 1996). In Leishmania-infected macrophages, there is a reduced expression of B7-1 even after lipopolysaccharide (LPS) stimulation, possibly mediated by parasite-induced prostaglandins (Saha, Das, Vohra, Ganguly, & Mishra, 1995). Similarly, during L. major infection B7-1 expression is down-regulated in epidermal cells from susceptible mice (Mbow, DeKrey, & Titus, 2001). Indirect evidence seems to support the notion that Leishmania increases the expression of B7-2 to favor the establishment of disease. In the same study by Mbow et al. (2001) B7-2 expression was higher in epidermal cells from susceptible mice than in resistant mice after L. major infection. In another study with human leishmaniasis caused by L. mexicana, B7-2 expression was higher in monocytes from patients with active DCL, while no change was observed in the expression of CD40 and B7-1 (Carrada et al., 2007). Furthermore, antibody blockade of B7-2, but not B7-1 led to an increased T cell response resulting in diminished parasite load (Murphy, Cotterell, Gorak, Engwerda, & Kaye, 1998; Murphy, Engwerda, Gorak, & Kaye, 1997). This is further supported by studies with L. major infected B7-1 knockout, B7-2 knockout, and B7-1/B7-2 double knockout BALB/c mice (Brown et al., 2002). Signaling via CD40-CD40L interactions, critical for the induction of anti-leishmanial responses (Campbell et al., 1996; Kamanaka et al., 1996), is also impaired by L. major (Awasthi et al., 2003).
2.5. Alteration of Host Cell Signaling
In order to survive inside macrophages armed with a host of microbicidal factors, Leishmania interferes with cell signaling cascades involved in their synthesis. One factor critical for early protective response against Leishmania (Murray & Nathan, 1999) is reactive oxygen species which is activated by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex (De Leo, Ulman, Davis, Jutila, & Quinn, 1996). In L. donovani-infected macrophages, phosphorylation of the p67 and p47 subunits of NADPH oxidase is blocked due to impaired protein kinase C (PKC) signaling (Bhattacharyya, Ghosh, Sen, Roy, & Majumdar, 2001) which favors parasite survival (Olivier, Brownsey, & Reiner, 1992; Turco et al., 1987). Leishmania promastigote LPG deactivates PKC by interfering with the binding of Ca2+ and diacyl glycerol to PKC and obstructing the insertion of PKC into the membrane (Descoteaux & Turco, 1999). Recent studies in L. mexicana-infected macrophages show that the ability of parasite-derived LPG to regulate the oxidative burst via PKC-α activity is related to the susceptible/resistant phenotypes observed genetic strains of mice (Delgado-Domínguez et al., 2010). Interestingly, amastigotes, devoid of LPG, have also shown similar attenuation in PKC activity (Olivier et al., 1992) suggesting the existence of LPG independent mechanisms such as Leishmania-induced ceramide (Ghosh et al., 2001) and IL-10 induction (Bhattacharyya, Ghosh, Jhonson, Bhattacharya, & Majumdar, 2001). GP63 which cleaves the PKC substrate, myristoylated alanine rich C kinase (MARCKS) and MARCKS-related protein (Olivier, Atayde, Isnard, Hassani, & Shio, 2012), and GIPL (Chawla & Vishwakarma, 2003) are virulence factors that are involved in the inhibition of PKC activity.
IFN-γ-mediated activation of the infected macrophage is critical for the destruction of intracellular Leishmania parasites. IFN-γ signals through the JAK/STAT pathway and is critical for the induction of nitric oxide, but this pathway is suppressed by Leishmania (Nandan & Reiner, 1995). Interaction of Leishmania promastigotes with macrophages activates SHP-1 which shows a strong and increased interaction with JAK2 resulting in its deactivation (Blanchette, Racette, Faure, Siminovitch, & Olivier, 1999). Further studies show that JAK2 inactivation by SHP-1 results in diminished nitric oxide production which favors parasite survival (Blanchette, Abu-Dayyeh, Hassani, Whitcombe, & Olivier, 2009). Further in Leishmania infected macrophages, STAT1 inactivation occurs and translocation of STAT1α to the nucleus is significantly diminished through a mechanism independent of SHP-1 activity (Forget, Gregory, & Olivier, 2005). In both L. major and L. mexicana infected macrophages treated with IFN-γ, phosphorylation of STAT1α is reduced, but dominant negative STAT1β phosphorylation is increased only in L. mexicana infected macrophages which points to species specific differences in the regulation of the JAK/STAT signaling pathway (Bhardwaj, Rosas, Lafuse, & Satoskar, 2005). Furthermore, quenching of membrane cholesterol has recently been shown to be a mechanism by which Leishmania impairs with IFN-γ signaling, and re-association of the signaling assembly could be restored by liposomal delivery of cholesterol together with IFN-γ (phospho-JAK1, JAK2, and STAT1) (Sen, Roy, Mukherjee, Mukhopadhyay, & Roy, 2011).
The MAPK signaling pathway (ERK1/2, JNK, and p38MAPK), required for the production of various effector molecules including cytokines and chemokines, is exploited by Leishmania as another immune evasive mechanism. This complex signaling pathway could either be activated or suppressed leading to the induction of gene products that favor parasite survival. While L. donovani inhibits EKR1/2, p38MAPK, and JNK activation in naïve macrophages leading to the suppression of pro-inflammatory cytokine production (Privé & Descoteaux, 2000), L. amazonensis activates ERK1/2 leading to the production of IL-10, a cytokine that contributes to parasite growth (Yang, Mosser, & Zhang, 2007). Mechanisms behind MAPK inactivation by Leishmania have been extensively studied. Studies with LPS activated macrophages show that Leishmania amastigotes can block the activation of EKR1/2 via the activation of ecto-protein phosphatase (Martiny, Meyer-Fernandes, de Souza, & Vannier-Santos, 1999). In PMA-activated macrophages, Leishmania amastigotes deactivated MAP kinase activity (ERK1/2), c-FOS and iNOS expression by activating cellular phosphotyrosine phosphatases (Nandan, Lo, & Reiner, 1999). Studies with SHP-1 deficient infected macrophages confirmed the pivotal role played by this phosphatase in the regulation of ERK1/2 signaling (Forget, Gregory, Whitcombe, & Olivier, 2006). Leishmania can also deactivate ERK1/2 by inducing ceramide generation in susceptible host macrophages (Ghosh et al., 2002). Interestingly, L. mexicana amastigotes block LPS induced IL-12 production by relying on their own cysteine proteinases for degrading the ERK1/2 and JNK but not p38MAPK (Cameron et al., 2004). Studies with CD40 ligand/antibody, showed that Leishmania interferes with the strength of CD40 cross-linking resulting in the reciprocal regulation of ERK1/2 and p38MAPK which governs the production of IL-10 and IL-12 in Leishmania-infected macrophages (Mathur, Awasthi, Wadhone, Ramanamurthy, & Saha, 2004). Moreover, Leishmania can redirect CD40-regulated immune responses via the reciprocal activation of MAPK phosphatases (MKP), MKP-1 and MKP-3 which reveal a novel parasite-devised immune evasion strategy (Srivastava, Sudan, & Saha, 2011).
Leishmania also regulates other important transcription factors such as AP-1 and NF-kB which have been shown to be important in immunity against the parasite. One study using RelA (NF-kB p65 subunit) knockout infected macrophages show higher intracellular parasite load and reduced NO levels compared to WT (Mise-Omata et al., 2009), establishing the importance of NF-kB in resistance to the disease. Leishmania specifically reduces the overall expression of RelA (Calegari-Silva et al., 2009). Leishmania mexicana promastigotes rely on the virulence factors GP63 and cysteine peptidase activity to cleave the RelA-p65 subunit into the smaller RelA-p35 subunit which activates specific chemokines that favor parasite multiplication (Abu-Dayyeh, Hassani, Westra, Mottram, & Olivier, 2010; Gregory, Godbout, Contreras, Forget, & Olivier, 2008). Leishmania mexicana amastigotes degrade the RelA-p65 subunit though cellular tyrosine phosphatases upon activation by parasite’s cysteine peptidase (Abu-Dayyeh et al., 2010). Leishmania major amastigotes favor IL-10 induction by selectively inhibiting p65–p50 complex thus allowing selective nuclear translocation of p50–p35 complex (Guizani-Tabbane, Ben-Aissa, Belghith, Sassi, & Dellagi, 2004). Leishmania also attenuates the activity of AP-1 (composed of dimmers of Fos and Jun family members) in infected macrophages in order to regulate the production of pro-inflammatory cytokines IL-1β, TNF-α, and IL-12 (Abu-Dayyeh et al., 2008; Contreras et al., 2010). Previous studies have shown that Leishmania interferes with the nuclear translocation of AP-1 which might be due to parasite-induced ceramide generation (Ghosh et al., 2002). Recent reports further show that Leishmania-derived GP63 enters the nucleus of the host macrophage to cleave c-Jun and c-Fos subunits of AP-1 rendering them inactive (Contreras et al., 2010). Thus by using multiple mechanisms Leishmania is able to interfere with the activity of transcription factors thereby subverting the host immune response.
Other host signaling targets of Leishmania include the mammalian mechanistic target of rapamycin (mTOR), which is cleaved by the Leishmania protease GP63 leading to the inhibition of mTOR complex 1 (mTORC1) and concomitant activation of 4E-BP1 to promote parasite survival (Jaramillo et al., 2011). Recently, L. donovani has been shown to utilize the mTOR signaling pathway to regulate IL-12 and IL-10 production in infected macrophages (Cheekatla, Aggarwal, & Naik, 2012).
2.6. Modulation of Cytokines and Chemokines
By interfering with host cell signaling pathways, many of which are still yet to be completely understood, various species of Leishmania possess the capability to modulate the cytokine profile of infected host cells to favor dissemination of the parasite and prevent its eradication. Pro-inflammatory cytokines (such as IL-12), which are essential for the generation of a successful immune response against Leishmania, are generally suppressed, facilitating a ‘silent entry’ of the parasite into host macrophages (Belkaid et al., 2000; McDowell & Sacks, 1999; Reiner, Zheng, Wang, Stowring, & Locksley, 1994; Weinheber, Wolfram, Harbecke, & Aebischer, 1998). Conversely, IL-10 which is essential for parasite survival and disease progression is induced by Leishmania in infected monocytes and macrophages (Chandra & Naik, 2008; Meddeb-Garnaoui, Zrelli, & Dellagi, 2009). Leishmania also expresses cytokine orthologs that modulate host immune cells. The human macrophage migration inhibitory factor ortholog, produced by L. major, has been shown to inhibit macrophage apoptosis and could contribute to parasite persistence and evasion of immune destruction (Kamir et al., 2008).
Chemokines and chemokine receptors play a major role in immunity to Leishmania by coordinating the recruitment and activation of anti-leishmanial immune cells (Oghumu, Lezama-Dávila, Isaac-Márquez, & Satoskar, 2010). Leishmania’s attempt to alter the chemokine expression profile in the infected tissue microenvironment will therefore contribute to their ability to evade the host’s immune system. For over a decade, Leishmania has been known to induce or inhibit the expression of chemokines to control recruitment of immune cells. In human cutaneous leishmaniasis, the diffuse form of the disease has been associated with lower levels of the macrophage chemotactic factor CCL2 (MCP-1), which is higher in the localized form of the disease (Ritter et al., 1996). Since CCL2 also induces antiparasitic activity in macrophages (Mannheimer, Hariprashad, Stoeckle, & Murray, 1996), inhibition of this chemokine by Leishmania could potentially facilitate its survival within the host. The Leishmania virulence factor LPG can also inhibit migration of monocytes across the endothelial wall by inhibiting the synthesis of CCL2 and the expression of cell surface adhesion molecules including E-selectin, ICAM-1, and VCAM-1 by endothelial cells (Lo et al., 1998). Expression of chemokine receptors CCR4 and CCR5 as well as integrin VLA-4 activity, all involved in macrophage adhesion, has also been shown to be inhibited during Leishmania infection (Pinheiro et al., 2006).
The ability of Leishmania to selectively recruit immune cells that will facilitate parasite survival was demonstrated by Katzman and Fowell (2008). This study showed that L. major can induce the expression of the Th2 attracting chemokine CCL7 at the dermal infection site, allowing the accumulation of IL-4 producing but not IFN-γ producing effector T cells. In contrast, ovalbumin with complete Freund’s adjuvant (OVA/CFA) immunized dermis permitted the accumulation of both populations of effector T cells, suggesting that L. major actively modulates the local tissue environment in an attempt to evade the hosts’ immune response. Leishmania promastigotes themselves secrete a chemotactic factor for PMNs, which serve as the host cell in the very early stages of infection (van Zandbergen, Hermann, Laufs, Solbach, & Laskay, 2002). At the same time they can induce the expression of IL-8 by these cells so as to recruit more PMNs which aid in the establishment of infection and proliferation of the parasite. On the other hand, they have the ability to inhibit the expression of IP-10, a chemokine that recruits and activates NK and Th1 cells, which are important in parasite eradication (van Zandbergen et al., 2002).
In a chronic murine VL model, L. donovani inhibited dendritic cell migration to T cell areas of the spleen due to a loss in CCR7 expression. Further, treatment of infected mice by adoptive transfer of CCR7 expressing dendritic cells significantly reduced parasite burdens (Ato, Stäger, Engwerda, & Kaye, 2002). Although mechanisms behind the process are still not fully understood, the induction or suppression of chemokine/chemokine receptor expression is a major way Leishmania evades the host immune system to favor establishment of disease.
2.7. Modification of T Cell Responses
Th1 cells play a vital role in the elimination of Leishmania through the secretion of IFN-γ and CD40L which activate macrophages to produce nitric oxide, a leishmanicidal factor. Recent studies have shown that some secreted factors from L. major possess immunosuppressive properties, could dampen lymphoproliferative capabilities and skew T cell polarization toward a susceptible Th2 phenotype (Tabatabaee, Abolhassani, Mahdavi, Nahrevanian, & Azadmanesh, 2011). Similarly, extracts of L. amazonensis, a causative agent of CL in the New World, promote a Th2 type immune response in the host, thereby enhancing infection (Silva et al., 2011).
The ability of Leishmania to induce the activity of regulatory T cells that suppress anti-Leishmania immune responses has been well established in cutaneous and visceral animal models of leishmaniasis (Belkaid, 2003; Belkaid, Piccirillo, et al., 2002; Mendez, Reckling, Piccirillo, Sacks, & Belkaid, 2004). This has also been shown in humans (Ganguly et al., 2010; Katara, Ansari, Verma, Ramesh, & Salotra, 2011; Rai et al., 2012). Tregs generally function as immune-regulators of cell-mediated immune responses, preventing pathology due to uncontrolled effector T cell activity. While this presents apparent advantages to the host, it could very well be capitalized by Leishmania, thereby preventing complete eradication of the parasite by Th1 cells. Tregs are retained at the site of infection where they secrete IL-10 and TGF-β which down regulate Th1 and macrophage activity, rendering the area of infection, an immune privileged site (Peters & Sacks, 2006).
In conclusion, current research that focuses on mechanisms of immune evasion by Leishmania could shed light on promising targets for therapeutic intervention. This is especially important as medication currently employed in the therapy of Leishmania presents problems due to toxicity, patient compliance, and drug resistance. Recent advances in the use of immuno-therapeutic approaches targeted at circumventing the parasites’ attempt at host immune evasion include the use of TLR agonists that induce strong cell mediated immune responses. The TLR 4 agonist monophosphoryl lipid A has been used in combination with Leishmania antigens or other agonists to protect against cutaneous or mucocutaneous Leishmania infection models (Aebischer et al., 2000; Coler & Reed, 2005; Raman et al., 2010) and some formulations are currently being used successfully in clinical trials (Llanos-Cuentas et al., 2010). Activation of the TLR2 pathway has been shown to reduce parasite burdens in L. donovani-infected macrophages (Bhattacharya et al., 2010). TLR2 agonist Pam3Cys and CPG-ODN 2006 have been used in combination with miltefosine in the immunotherapy of experimental visceral leishmaniasis (Gupta, Sane, Shakya, Vishwakarma, & Haq, 2011; Shakya, Sane, Shankar, & Gupta, 2011).
Other promising immuno-therapeutic approaches involve the use of chemokines or chemokine receptor agonists (Gupta, Majumdar, et al., 2011), blockade of co-stimulatory molecules (Murphy et al., 1998, 1997), reconstitution of membrane cholesterol to facilitate re-association of membrane signaling assemblies (Sen et al., 2011), and inhibitors of cell signaling molecules (Cheekatla et al., 2012; Cummings et al., 2012), all mechanisms of which are regulated by Leishmania to evade or exploit host immune responses. While this presents an area for extensive future research, current advances have made significant impact and present weighty implications in the management of leishmaniasis.
Acknowledgments
This work was supported by National Institutes of Health grants R03AI090231, RC4AI092624, R34AI100789, R21AT004160 and R03CA164399 awarded to A.R.S and National Institute of Dental and Craniofacial Research Training Grant T32DE014320 awarded to S.O.
References
- Abu-Dayyeh I, Hassani K, Westra ER, Mottram JC, Olivier M. Comparative study of the ability of Leishmania mexicana promastigotes and amastigotes to alter macrophage signaling and functions. Infection and Immunity. 2010;78:2438–2445. doi: 10.1128/IAI.00812-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abu-Dayyeh I, Shio MT, Sato S, Akira S, Cousineau B, Olivier M. Leishmania-induced IRAK-1 inactivation is mediated by SHP-1 interacting with an evolutionarily conserved KTIM motif. PLoS Neglected Tropical Diseases. 2008;2:e305. doi: 10.1371/journal.pntd.0000305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aebischer T, Wolfram M, Patzer SI, Ilg T, Wiese M, Overath P. Subunit vaccination of mice against new world cutaneous leishmaniasis: comparison of three proteins expressed in amastigotes and six adjuvants. Infection and Immunity. 2000;68:1328–1336. doi: 10.1128/iai.68.3.1328-1336.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Afonso L, Borges VM, Cruz H, Ribeiro-Gomes FL, DosReis GA, Dutra AN, et al. Interactions with apoptotic but not with necrotic neutrophils increase parasite burden in human macrophages infected with Leishmania amazonensis. Journal of Leukocyte Biology. 2008;84:389–396. doi: 10.1189/jlb.0108018. [DOI] [PubMed] [Google Scholar]
- Alexander J, Satoskar AR, Russell DG. Leishmania species: models of intracellular parasitism. Journal of Cell Science. 1999;112(Pt 18):2993–3002. doi: 10.1242/jcs.112.18.2993. [DOI] [PubMed] [Google Scholar]
- Ali HZ, Harding CR, Denny PW. Endocytosis and sphingolipid scavenging in Leishmania mexicana amastigotes. Biochemistry Research International. 2012;2012:691363. doi: 10.1155/2012/691363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amprey JL, Im JS, Turco SJ, Murray HW, Illarionov PA, Besra GS, et al. A subset of liver NK T cells is activated during Leishmania donovani infection by CD1d-bound lipophosphoglycan. Journal of Experimental Medicine. 2004;200:895–904. doi: 10.1084/jem.20040704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ato M, Stäger S, Engwerda CR, Kaye PM. Defective CCR7 expression on dendritic cells contributes to the development of visceral leishmaniasis. Nature Immunology. 2002;3:1185–1191. doi: 10.1038/ni861. [DOI] [PubMed] [Google Scholar]
- Awasthi A, Mathur R, Khan A, Joshi BN, Jain N, Sawant S, et al. CD40 signaling is impaired in L. major-infected macrophages and is rescued by a p38MAPK activator establishing a host-protective memory T cell response. Journal of Experimental Medicine. 2003;197:1037–1043. doi: 10.1084/jem.20022033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Awasthi A, Mathur RK, Saha B. Immune response to Leishmania infection. Indian Journal of Medical Research. 2004;119:238–258. [PubMed] [Google Scholar]
- Bankoti R, Gupta K, Levchenko A, Stäger S. Marginal zone B cells regulate antigen-specific T cell responses during infection. Journal of Immunology. 2012;188:3961–3971. doi: 10.4049/jimmunol.1102880. [DOI] [PubMed] [Google Scholar]
- Belkaid Y. The role of CD4(+)CD25(+) regulatory T cells in Leishmania infection. Expert Opinion on Biological Therapy. 2003;3:875–885. doi: 10.1517/14712598.3.6.875. [DOI] [PubMed] [Google Scholar]
- Belkaid Y, Mendez S, Lira R, Kadambi N, Milon G, Sacks D. A natural model of Leishmania major infection reveals a prolonged “silent” phase of parasite amplification in the skin before the onset of lesion formation and immunity. Journal of Immunology. 2000;165:969–977. doi: 10.4049/jimmunol.165.2.969. [DOI] [PubMed] [Google Scholar]
- Belkaid Y, Piccirillo CA, Mendez S, Shevach EM, Sacks DL. CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity. Nature. 2002;420:502–507. doi: 10.1038/nature01152. [DOI] [PubMed] [Google Scholar]
- Belkaid Y, Von Stebut E, Mendez S, Lira R, Caler E, Bertholet S, et al. CD8+ T cells are required for primary immunity in C57BL/6 mice following low-dose, intradermal challenge with Leishmania major. Journal of Immunology. 2002;168:3992–4000. doi: 10.4049/jimmunol.168.8.3992. [DOI] [PubMed] [Google Scholar]
- Bhardwaj N, Rosas LE, Lafuse WP, Satoskar AR. Leishmania inhibits STAT1-mediated IFN-gamma signaling in macrophages: increased tyrosine phosphorylation of dominant negative STAT1beta by Leishmania mexicana. International Journal of Parasitology. 2005;35:75–82. doi: 10.1016/j.ijpara.2004.10.018. [DOI] [PubMed] [Google Scholar]
- Bhattacharya P, Bhattacharjee S, Gupta G, Majumder S, Adhikari A, Mukherjee A, et al. Arabinosylated lipoarabinomannan-mediated protection in visceral leishmaniasis through up-regulation of toll-like receptor 2 signaling: an immunoprophylactic approach. Journal of Infectious Diseases. 2010;202:145–155. doi: 10.1086/653210. [DOI] [PubMed] [Google Scholar]
- Bhattacharyya S, Ghosh S, Jhonson PL, Bhattacharya SK, Majumdar S. Immunomodulatory role of interleukin-10 in visceral leishmaniasis: defective activation of protein kinase C-mediated signal transduction events. Infection and Immunity. 2001;69:1499–1507. doi: 10.1128/IAI.69.3.1499-1507.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhattacharyya S, Ghosh S, Sen P, Roy S, Majumdar S. Selective impairment of protein kinase C isotypes in murine macrophage by Leishmania donovani. Molecular and Cellular Biochemistry. 2001;216:47–57. doi: 10.1023/a:1011048210691. [DOI] [PubMed] [Google Scholar]
- 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. Journal of Experimental Medicine. 1985;162:324–331. doi: 10.1084/jem.162.1.324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blanchette J, Abu-Dayyeh I, Hassani K, Whitcombe L, Olivier M. Regulation of macrophage nitric oxide production by the protein tyrosine phosphatase Src homology 2 domain phosphotyrosine phosphatase 1 (SHP-1) Immunology. 2009;127:123–133. doi: 10.1111/j.1365-2567.2008.02929.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blanchette J, Racette N, Faure R, Siminovitch KA, Olivier M. Leishmania-induced increases in activation of macrophage SHP-1 tyrosine phosphatase are associated with impaired IFN-gamma-triggered JAK2 activation. European Journal of Immunology. 1999;29:3737–3744. doi: 10.1002/(SICI)1521-4141(199911)29:11<3737::AID-IMMU3737>3.0.CO;2-S. [DOI] [PubMed] [Google Scholar]
- Bogdan C, Gessner A, Solbach W, Röllinghoff M. Invasion, control and persistence of Leishmania parasites. Current Opinion in Immunology. 1996;8:517–525. doi: 10.1016/s0952-7915(96)80040-9. [DOI] [PubMed] [Google Scholar]
- Brittingham A, Morrison CJ, McMaster WR, McGwire BS, Chang KP, Mosser DM. Role of the Leishmania surface protease gp63 in complement fixation, cell adhesion, and resistance to complement-mediated lysis. Journal of Immunology. 1995;155:3102–3111. [PubMed] [Google Scholar]
- Brown JA, Greenwald RJ, Scott S, Schweitzer AN, Satoskar AR, Chung C, et al. T helper differentiation in resistant and susceptible B7-deficient mice infected with Leishmania major. European Journal of Immunology. 2002;32:1764–1772. doi: 10.1002/1521-4141(200206)32:6<1764::AID-IMMU1764>3.0.CO;2-V. [DOI] [PubMed] [Google Scholar]
- Calegari-Silva TC, Pereira RM, De-Melo LD, Saraiva EM, Soares DC, Bellio M, et al. NF-kappaB-mediated repression of iNOS expression in Leishmania amazonensis macrophage infection. Immunology Letters. 2009;127:19–26. doi: 10.1016/j.imlet.2009.08.009. [DOI] [PubMed] [Google Scholar]
- Cameron P, McGachy A, Anderson M, Paul A, Coombs GH, Mottram JC, et al. Inhibition of lipopolysaccharide-induced macrophage IL-12 production by Leishmania mexicana amastigotes: the role of cysteine peptidases and the NF-kappaB signaling pathway. Journal of Immunology. 2004;173:3297–3304. doi: 10.4049/jimmunol.173.5.3297. [DOI] [PubMed] [Google Scholar]
- Campbell KA, Ovendale PJ, Kennedy MK, Fanslow WC, Reed SG, Maliszewski CR. CD40 ligand is required for protective cell-mediated immunity to Leishmania major. Immunity. 1996;4:283–289. doi: 10.1016/s1074-7613(00)80436-7. [DOI] [PubMed] [Google Scholar]
- Camussi G, Bussolino F, Salvidio G, Baglioni C. Tumor necrosis factor/cachectin stimulates peritoneal macrophages, polymorphonuclear neutrophils, and vascular endothelial cells to synthesize and release platelet-activating factor. Journal of Experimental Medicine. 1987;166:1390–1404. doi: 10.1084/jem.166.5.1390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carrada G, Cañeda C, Salaiza N, Delgado J, Ruiz A, Sanchez B, et al. Monocyte cytokine and costimulatory molecule expression in patients infected with Leishmania mexicana. Parasite Immunology. 2007;29:117–126. doi: 10.1111/j.1365-3024.2006.00924.x. [DOI] [PubMed] [Google Scholar]
- Carter CR, Whitcomb JP, Campbell JA, Mukbel RM, McDowell MA. Complement receptor 3 deficiency influences lesion progression during Leishmania major infection in BALB/c mice. Infection and Immunity. 2009;77:5668–5675. doi: 10.1128/IAI.00802-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chakkalath HR, Theodos CM, Markowitz JS, Grusby MJ, Glimcher LH, Titus RG. Class II major histocompatibility complex-deficient mice initially control an infection with Leishmania major but succumb to the disease. Journal of Infectious Diseases. 1995;171:1302–1308. doi: 10.1093/infdis/171.5.1302. [DOI] [PubMed] [Google Scholar]
- Chakraborty D, Banerjee S, Sen A, Banerjee KK, Das P, Roy S. Leishmania donovani affects antigen presentation of macrophage by disrupting lipid rafts. Journal of Immunology. 2005;175:3214–3224. doi: 10.4049/jimmunol.175.5.3214. [DOI] [PubMed] [Google Scholar]
- Chandra D, Naik S. Leishmania donovani infection down-regulates TLR2-stimulated IL-12p40 and activates IL-10 in cells of macrophage/monocytic lineage by modulating MAPK pathways through a contact-dependent mechanism. Clinical and Experimental Immunology. 2008;154:224–234. doi: 10.1111/j.1365-2249.2008.03741.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Charmoy M, Megnekou R, Allenbach C, Zweifel C, Perez C, Monnat K, et al. Leishmania major induces distinct neutrophil phenotypes in mice that are resistant or susceptible to infection. Journal of Leukocyte Biology. 2007;82:288–299. doi: 10.1189/jlb.0706440. [DOI] [PubMed] [Google Scholar]
- Chawla M, Vishwakarma RA. Alkylacylglycerolipid domain of GPI molecules of Leishmania is responsible for inhibition of PKC-mediated c-fos expression. Journal of Lipid Research. 2003;44:594–600. doi: 10.1194/jlr.M200296-JLR200. [DOI] [PubMed] [Google Scholar]
- Cheekatla SS, Aggarwal A, Naik S. mTOR signaling pathway regulates the IL-12/IL-10 axis in Leishmania donovani infection. Medical Microbiology and Immunology. 2012;201:37–46. doi: 10.1007/s00430-011-0202-5. [DOI] [PubMed] [Google Scholar]
- Coler RN, Reed SG. Second-generation vaccines against leishmaniasis. Trends in Parasitology. 2005;21:244–249. doi: 10.1016/j.pt.2005.03.006. [DOI] [PubMed] [Google Scholar]
- Contreras I, Gómez MA, Nguyen O, Shio MT, McMaster RW, Olivier M. Leishmania-induced inactivation of the macrophage transcription factor AP-1 is mediated by the parasite metalloprotease GP63. PLoS Pathogens. 2010;6:e1001148. doi: 10.1371/journal.ppat.1001148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Courret N, Prina E, Mougneau E, Saraiva EM, Sacks DL, Glaichenhaus N, et al. Presentation of the Leishmania antigen LACK by infected macrophages is dependent upon the virulence of the phagocytosed parasites. European Journal of Immunology. 1999;29:762–773. doi: 10.1002/(SICI)1521-4141(199903)29:03<762::AID-IMMU762>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
- Culley FJ, Harris RA, Kaye PM, McAdam KP, Raynes JG. C-reactive protein binds to a novel ligand on Leishmania donovani and increases uptake into human macrophages. Journal of Immunology. 1996;156:4691–4696. [PubMed] [Google Scholar]
- Cummings HE, Barbi J, Reville P, Oghumu S, Zorko N, Sarkar A, et al. Critical role for phosphoinositide 3-kinase gamma in parasite invasion and disease progression of cutaneous leishmaniasis. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:1251–1256. doi: 10.1073/pnas.1110339109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Da Silva RP, Hall BF, Joiner KA, Sacks DL. CR1, the C3b receptor, mediates binding of infective Leishmania major metacyclic promastigotes to human macrophages. Journal of Immunology. 1989;143:617–622. [PubMed] [Google Scholar]
- Das S, Pandey K, Kumar A, Sardar AH, Purkait B, Kumar M, et al. TGF-β(1) re-programs TLR4 signaling in L. donovani infection: enhancement of SHP-1 and ubiquitin-editing enzyme A20. Immunology and Cell Biology. 2012;90:640–654. doi: 10.1038/icb.2011.80. [DOI] [PubMed] [Google Scholar]
- De Leo FR, Ulman KV, Davis AR, Jutila KL, Quinn MT. Assembly of the human neutrophil NADPH oxidase involves binding of p67phox and flavocyto-chrome b to a common functional domain in p47phox. Journal of Biological Chemistry. 1996;271:17013–17020. doi: 10.1074/jbc.271.29.17013. [DOI] [PubMed] [Google Scholar]
- De Souza Leao S, Lang T, Prina E, Hellio R, Antoine JC. Intracellular Leishmania amazonensis amastigotes internalize and degrade MHC class II molecules of their host cells. Journal of Cell Science. 1995;108(Pt 10):3219–3231. doi: 10.1242/jcs.108.10.3219. [DOI] [PubMed] [Google Scholar]
- Delgado-Domínguez J, González-Aguilar H, Aguirre-García M, Gutiérrez-Kobeh L, Berzunza-Cruz M, Ruiz-Remigio A, et al. Leishmania mexicana lipophospho-glycan differentially regulates PKCalpha-induced oxidative burst in macrophages of BALB/c and C57BL/6 mice. Parasite Immunology. 2010;32:440–449. doi: 10.1111/j.1365-3024.2010.01205.x. [DOI] [PubMed] [Google Scholar]
- Descoteaux A, Turco SJ. Glycoconjugates in Leishmania infectivity. Biochimica et Biophysica Acta. 1999;1455:341–352. doi: 10.1016/s0925-4439(99)00065-4. [DOI] [PubMed] [Google Scholar]
- Desjeux P. Leishmaniasis: current situation and new perspectives. Comparative Immunology. Microbiology and Infectious Diseases. 2004;27:305–318. doi: 10.1016/j.cimid.2004.03.004. [DOI] [PubMed] [Google Scholar]
- Diefenbach A, Schindler H, Donhauser N, Lorenz E, Laskay T, MacMicking J, et al. Type 1 interferon (IFNalpha/beta) and type 2 nitric oxide synthase regulate the innate immune response to a protozoan parasite. Immunity. 1998;8:77–87. doi: 10.1016/s1074-7613(00)80460-4. [DOI] [PubMed] [Google Scholar]
- Erb K, Blank C, Ritter U, Bluethmann H, Moll H. Leishmania major infection in major histocompatibility complex class II-deficient mice: CD8+ T cells do not mediate a protective immune response. Immunobiology. 1996;195:243–260. doi: 10.1016/S0171-2985(96)80043-X. [DOI] [PubMed] [Google Scholar]
- Faria MS, Reis FC, Azevedo-Pereira RL, Morrison LS, Mottram JC, Lima AP. Leishmania inhibitor of serine peptidase 2 prevents TLR4 activation by neutrophil elastase promoting parasite survival in murine macrophages. Journal of Immunology. 2011;186:411–422. doi: 10.4049/jimmunol.1002175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Faria MS, Reis FC, Lima AP. Toll-like receptors in Leishmania infections: guardians or promoters? Journal of Parasitology Research. 2012;2012:930257. doi: 10.1155/2012/930257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Filardy AA, Pires DR, Nunes MP, Takiya CM, Freire-de-Lima CG, Ribeiro-Gomes FL, et al. Proinflammatory clearance of apoptotic neutrophils induces an IL-12(low)IL-10(high) regulatory phenotype in macrophages. Journal of Immunology. 2010;185:2044–2050. doi: 10.4049/jimmunol.1000017. [DOI] [PubMed] [Google Scholar]
- Flandin JF, Chano F, Descoteaux A. RNA interference reveals a role for TLR2 and TLR3 in the recognition of Leishmania donovani promastigotes by interferon-gamma-primed macrophages. European Journal of Immunology. 2006;36:411–420. doi: 10.1002/eji.200535079. [DOI] [PubMed] [Google Scholar]
- Forget G, Gregory DJ, Olivier M. Proteasome-mediated degradation of STAT-1alpha following infection of macrophages with Leishmania donovani. Journal of Biological Chemistry. 2005;280:30542–30549. doi: 10.1074/jbc.M414126200. [DOI] [PubMed] [Google Scholar]
- Forget G, Gregory DJ, Whitcombe LA, Olivier M. Role of host protein tyrosine phosphatase SHP-1 in Leishmania donovani-induced inhibition of nitric oxide production. Infection and Immunity. 2006;74:6272–6279. doi: 10.1128/IAI.00853-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Freitas Balanco JM, Moreira ME, Bonomo A, Bozza PT, Amarante-Mendes G, Pirmez C, et al. Apoptotic mimicry by an obligate intracellular parasite down-regulates macrophage microbicidal activity. Current Biology. 2001;11:1870–1873. doi: 10.1016/s0960-9822(01)00563-2. [DOI] [PubMed] [Google Scholar]
- Fruth U, Solioz N, Louis JA. Leishmania major interferes with antigen presentation by infected macrophages. Journal of Immunology. 1993;150:1857–1864. [PubMed] [Google Scholar]
- Ganguly S, Mukhopadhyay D, Das NK, Chaduvula M, Sadhu S, Chatterjee U, et al. Enhanced lesional Foxp3 expression and peripheral anergic lymphocytes indicate a role for regulatory T cells in Indian post-kala-azar dermal leishmaniasis. Journal of Investigative Dermatology. 2010;130:1013–1022. doi: 10.1038/jid.2009.393. [DOI] [PubMed] [Google Scholar]
- Gaur U, Roberts SC, Dalvi RP, Corraliza I, Ullman B, Wilson ME. An effect of parasite-encoded arginase on the outcome of murine cutaneous leishmaniasis. Journal of Immunology. 2007;179:8446–8453. doi: 10.4049/jimmunol.179.12.8446. [DOI] [PubMed] [Google Scholar]
- Ghosh S, Bhattacharyya S, Das S, Raha S, Maulik N, Das DK, et al. Generation of ceramide in murine macrophages infected with Leishmania donovani alters macrophage signaling events and aids intracellular parasitic survival. Molecular and Cellular Biochemistry. 2001;223:47–60. doi: 10.1023/a:1017996609928. [DOI] [PubMed] [Google Scholar]
- Ghosh S, Bhattacharyya S, Sirkar M, Sa GS, Das T, Majumdar D, et al. Leishmania donovani suppresses activated protein 1 and NF-kappaB activation in host macrophages via ceramide generation: involvement of extracellular signal-regulated kinase. Infection and Immunity. 2002;70:6828–6838. doi: 10.1128/IAI.70.12.6828-6838.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gomes-Pereira S, Rodrigues OR, Rolão N, Almeida PD, Santos-Gomes GM. Hepatic cellular immune responses in mice with “cure” and “non-cure” phenotype to Leishmania infantum infection: importance of CD8+ T cells and TGF-beta production. FEMS Immunology and Medical Microbiology. 2004;41:59–68. doi: 10.1016/j.femsim.2004.01.003. [DOI] [PubMed] [Google Scholar]
- Gregory DJ, Godbout M, Contreras I, Forget G, Olivier M. A novel form of NF-kappaB is induced by Leishmania infection: involvement in macrophage gene expression. European Journal of Immunology. 2008;38:1071–1081. doi: 10.1002/eji.200737586. [DOI] [PubMed] [Google Scholar]
- Guimarães-Costa AB, Nascimento MT, Froment GS, Soares RP, Morgado FN, Conceição-Silva F, et al. Leishmania amazonensis promastigotes induce and are killed by neutrophil extracellular traps. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:6748–6753. doi: 10.1073/pnas.0900226106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guizani-Tabbane L, Ben-Aissa K, Belghith M, Sassi A, Dellagi K. Leishmania major amastigotes induce p50/c-Rel NF-kappa B transcription factor in human macrophages: involvement in cytokine synthesis. Infection and Immunity. 2004;72:2582–2589. doi: 10.1128/IAI.72.5.2582-2589.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gupta G, Majumdar S, Adhikari A, Bhattacharya P, Mukherjee AK, Majumdar SB. Treatment with IP-10 induces host-protective immune response by regulating the T regulatory cell functioning in Leishmania donovani-infected mice. Medical Microbiology and Immunology. 2011;200:241–253. doi: 10.1007/s00430-011-0197-y. [DOI] [PubMed] [Google Scholar]
- Gupta S, Sane SA, Shakya N, Vishwakarma P, Haq W. CpG oligodeoxy-nucleotide 2006 and miltefosine, a potential combination for treatment of experimental visceral leishmaniasis. Antimicrobial Agents and Chemotherapy. 2011;55:3461–3464. doi: 10.1128/AAC.00137-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guy RA, Belosevic M. Comparison of receptors required for entry of Leishmania major amastigotes into macrophages. Infection and Immunity. 1993;61:1553–1558. doi: 10.1128/iai.61.4.1553-1558.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hartley MA, Ronet C, Zangger H, Beverley SM, Fasel N. Leishmania RNA virus: when the host pays the toll. Frontiers in Cellular and Infection Microbiology. 2012;2:99. doi: 10.3389/fcimb.2012.00099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hermoso T, Fishelson Z, Becker SI, Hirschberg K, Jaffe CL. Leishmanial protein kinases phosphorylate components of the complement system. EMBO Journal. 1991;10:4061–4067. doi: 10.1002/j.1460-2075.1991.tb04982.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holm A, Tejle K, Magnusson KE, Descoteaux A, Rasmusson B. Leishmania donovani lipophosphoglycan causes periphagosomal actin accumulation: correlation with impaired translocation of PKCalpha and defective phagosome maturation. Cellular Microbiology. 2001;3:439–447. doi: 10.1046/j.1462-5822.2001.00127.x. [DOI] [PubMed] [Google Scholar]
- Hoover DL, Berger M, Nacy CA, Hockmeyer WT, Meltzer MS. Killing of Leishmania tropica amastigotes by factors in normal human serum. Journal of Immunology. 1984;132:893–897. [PubMed] [Google Scholar]
- Huber M, Timms E, Mak TW, Röllinghoff M, Lohoff M. Effective and long-lasting immunity against the parasite Leishmania major in CD8-deficient mice. Infection and Immunity. 1998;66:3968–3970. doi: 10.1128/iai.66.8.3968-3970.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huynh C, Andrews NW. Iron acquisition within host cells and the pathogenicity of Leishmania. Cellular Microbiology. 2008;10:293–300. doi: 10.1111/j.1462-5822.2007.01095.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iniesta V, Gómez-Nieto LC, Corraliza I. The inhibition of arginase by N(omega)-hydroxy-l-arginine controls the growth of Leishmania inside macrophages. Journal of Experimental Medicine. 2001;193:777–784. doi: 10.1084/jem.193.6.777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishikawa H, Hisaeda H, Taniguchi M, Nakayama T, Sakai T, Maekawa Y, et al. CD4(+) v(alpha)14 NKT cells play a crucial role in an early stage of protective immunity against infection with Leishmania major. International Immunology. 2000;12:1267–1274. doi: 10.1093/intimm/12.9.1267. [DOI] [PubMed] [Google Scholar]
- Ives A, Ronet C, Prevel F, Ruzzante G, Fuertes-Marraco S, Schutz F, et al. Leishmania RNA virus controls the severity of mucocutaneous leishmaniasis. Science. 2011;331:775–778. doi: 10.1126/science.1199326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaramillo M, Gomez MA, Larsson O, Shio MT, Topisirovic I, Contreras I, et al. Leishmania repression of host translation through mTOR cleavage is required for parasite survival and infection. Cell Host and Microbe. 2011;9:331–341. doi: 10.1016/j.chom.2011.03.008. [DOI] [PubMed] [Google Scholar]
- Kamanaka M, Yu P, Yasui T, Yoshida K, Kawabe T, Horii T, et al. Protective role of CD40 in Leishmania major infection at two distinct phases of cell-mediated immunity. Immunity. 1996;4:275–281. doi: 10.1016/s1074-7613(00)80435-5. [DOI] [PubMed] [Google Scholar]
- Kamir D, Zierow S, Leng L, Cho Y, Diaz Y, Griffith J, et al. A Leishmania ortho-log of macrophage migration inhibitory factor modulates host macrophage responses. Journal of Immunology. 2008;180:8250–8261. doi: 10.4049/jimmunol.180.12.8250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kane MM, Mosser DM. Leishmania parasites and their ploys to disrupt macrophage activation. Current Opinion in Hematology. 2000;7:26–31. doi: 10.1097/00062752-200001000-00006. [DOI] [PubMed] [Google Scholar]
- Karmakar S, Bhaumik SK, Paul J, De T. TLR4 and NKT cell synergy in immunotherapy against visceral leishmaniasis. PLoS Pathogens. 2012;8:e1002646. doi: 10.1371/journal.ppat.1002646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karmakar S, Paul J, De T. Leishmania donovani glycosphingolipid facilitates antigen presentation by inducing relocation of CD1d into lipid rafts in infected macrophages. European Journal of Immunology. 2011;41:1376–1387. doi: 10.1002/eji.201040981. [DOI] [PubMed] [Google Scholar]
- Katara GK, Ansari NA, Verma S, Ramesh V, Salotra P. Foxp3 and IL-10 expression correlates with parasite burden in lesional tissues of post kala azar dermal leishmaniasis (PKDL) patients. PLoS Neglected Tropical Diseases. 2011;5:e1171. doi: 10.1371/journal.pntd.0001171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Katzman SD, Fowell DJ. Pathogen-imposed skewing of mouse chemokine and cytokine expression at the infected tissue site. Journal of Clinical Investigation. 2008;118:801–811. doi: 10.1172/JCI33174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kautz-Neu K, Noordegraaf M, Dinges S, Bennett CL, John D, Clausen BE, et al. Langerhans cells are negative regulators of the anti-Leishmania response. Journal of Experimental Medicine. 2011;208:885–891. doi: 10.1084/jem.20102318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kavoosi G, Ardestani SK, Kariminia A. The involvement of TLR2 in cytokine and reactive oxygen species (ROS) production by PBMCs in response to Leishmania major phosphoglycans (PGs) Parasitology. 2009;136:1193–1199. doi: 10.1017/S0031182009990473. [DOI] [PubMed] [Google Scholar]
- Kavoosi G, Ardestani SK, Kariminia A, Alimohammadian MH. Leishmania major lipophosphoglycan: discrepancy in toll-like receptor signaling. Experimental Parasitology. 2010;124:214–218. doi: 10.1016/j.exppara.2009.09.017. [DOI] [PubMed] [Google Scholar]
- Kaye P, Scott P. Leishmaniasis: complexity at the host-pathogen interface. Nature Reviews Microbiology. 2011;9:604–615. doi: 10.1038/nrmicro2608. [DOI] [PubMed] [Google Scholar]
- Khouri R, Bafica A, Silva MaP, Noronha A, Kolb JP, Wietzerbin J, et al. IFN-beta impairs superoxide-dependent parasite killing in human macrophages: evidence for a deleterious role of SOD1 in cutaneous leishmaniasis. Journal of Immunology. 2009;182:2525–2531. doi: 10.4049/jimmunol.0802860. [DOI] [PubMed] [Google Scholar]
- Kima PE, Soong L, Chicharro C, Ruddle NH, McMahon-Pratt D. Leishmania-infected macrophages sequester endogenously synthesized parasite antigens from presentation to CD4+ T cells. European Journal of Immunology. 1996;26:3163–3169. doi: 10.1002/eji.1830261249. [DOI] [PubMed] [Google Scholar]
- Kirkpatrick CE, Farrell JP. Leishmaniasis in beige mice. Infection and Immunity. 1982;38:1208–1216. doi: 10.1128/iai.38.3.1208-1216.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kopf M, Brombacher F, Kohler G, Kienzle G, Widmann KH, Lefrang K, et al. IL-4-deficient Balb/c mice resist infection with Leishmania major. Journal of Experimental Medicine. 1996;184:1127–1136. doi: 10.1084/jem.184.3.1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Journal of Leukocyte Biology. 2004;76:48–57. doi: 10.1189/jlb.1003484. [DOI] [PubMed] [Google Scholar]
- Kropf P, Freudenberg MA, Modolell M, Price HP, Herath S, Antoniazi S, et al. Toll-like receptor 4 contributes to efficient control of infection with the protozoan parasite Leishmania major. Infection and Immunity. 2004;72:1920–1928. doi: 10.1128/IAI.72.4.1920-1928.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kropf P, Herath S, Weber V, Modolell M, Müller I. Factors influencing Leishmania major infection in IL-4-deficient BALB/c mice. Parasite Immunology. 2003;25:439–447. doi: 10.1111/j.1365-3024.2003.00655.x. [DOI] [PubMed] [Google Scholar]
- Kushawaha PK, Gupta R, Sundar S, Sahasrabuddhe AA, Dube A. Elongation factor-2, a Th1 stimulatory protein of Leishmania donovani, generates strong IFN-γ and IL-12 response in cured Leishmania-infected patients/hamsters and protects hamsters against Leishmania challenge. Journal of Immunology. 2011;187:6417–6427. doi: 10.4049/jimmunol.1102081. [DOI] [PubMed] [Google Scholar]
- Lang T, de Chastellier C, Frehel C, Hellio R, Metezeau P, Leao SeS, et al. Distribution of MHC class I and of MHC class II molecules in macrophages infected with Leishmania amazonensis. Journal of Cell Science. 1994;107(Pt 1):69–82. doi: 10.1242/jcs.107.1.69. [DOI] [PubMed] [Google Scholar]
- Laskay T, Diefenbach A, Röllinghoff M, Solbach W. Early parasite containment is decisive for resistance to Leishmania major infection. European Journal of Immunology. 1995;25:2220–2227. doi: 10.1002/eji.1830250816. [DOI] [PubMed] [Google Scholar]
- Lieke T, Nylén S, Eidsmo L, Schmetz C, Berg L, Akuffo H. The interplay between Leishmania promastigotes and human natural killer cells in vitro leads to direct lysis of Leishmania by NK cells and modulation of NK cell activity by Leishmania pro-mastigotes. Parasitology. 2011;138:1898–1909. doi: 10.1017/S0031182011001363. [DOI] [PubMed] [Google Scholar]
- Liu D, Zhang T, Marshall AJ, Okkenhaug K, Vanhaesebroeck B, Uzonna JE. The p110delta isoform of phosphatidylinositol 3-kinase controls susceptibility to Leishmania major by regulating expansion and tissue homing of regulatory T cells. Journal of Immunology. 2009;183:1921–1933. doi: 10.4049/jimmunol.0901099. [DOI] [PubMed] [Google Scholar]
- Llanos-Cuentas A, Calderon W, Cruz M, Ashman JA, Alves FP, Coler RN, et al. A clinical trial to evaluate the safety and immunogenicity of the LEISH-F1+MPL-SE vaccine when used in combination with sodium stibogluconate for the treatment of mucosal leishmaniasis. Vaccine. 2010;28:7427–7435. doi: 10.1016/j.vaccine.2010.08.092. [DOI] [PubMed] [Google Scholar]
- Lo SK, Bovis L, Matura R, Zhu B, He S, Lum H, et al. Leishmania lipophosphoglycan reduces monocyte transendothelial migration: modulation of cell adhesion molecules, intercellular junctional proteins, and chemoattractants. Journal of Immunology. 1998;160:1857–1865. [PubMed] [Google Scholar]
- Locksley RM, Reiner SL, Hatam F, Littman DR, Killeen N. Helper T cells without CD4: control of leishmaniasis in CD4-deficient mice. Science. 1993;261:1448–1451. doi: 10.1126/science.8367726. [DOI] [PubMed] [Google Scholar]
- Mannheimer SB, Hariprashad J, Stoeckle MY, Murray HW. Induction of macrophage antiprotozoal activity by monocyte chemotactic and activating factor. FEMS Immunology and Medical Microbiology. 1996;14:59–61. doi: 10.1111/j.1574-695X.1996.tb00268.x. [DOI] [PubMed] [Google Scholar]
- Marth T, Kelsall BL. Regulation of interleukin-12 by complement receptor 3 signaling. Journal of Experimental Medicine. 1997;185:1987–1995. doi: 10.1084/jem.185.11.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martiny A, Meyer-Fernandes JR, de Souza W, Vannier-Santos MA. Altered tyrosine phosphorylation of ERK1 MAP kinase and other macrophage molecules caused by Leishmania amastigotes. Molecular and Biochemical Parasitology. 1999;102:1–12. doi: 10.1016/s0166-6851(99)00067-5. [DOI] [PubMed] [Google Scholar]
- Mary C, Auriault V, Faugère B, Dessein AJ. Control of Leishmania infantum infection is associated with CD8(+) and gamma interferon- and interleukin-5-producing CD4(+) antigen-specific T cells. Infection and Immunity. 1999;67:5559–5566. doi: 10.1128/iai.67.11.5559-5566.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mathur RK, Awasthi A, Wadhone P, Ramanamurthy B, Saha B. Reciprocal CD40 signals through p38MAPK and ERK-1/2 induce counteracting immune responses. Nature Medicine. 2004;10:540–544. doi: 10.1038/nm1045. [DOI] [PubMed] [Google Scholar]
- Mattner J, Donhauser N, Werner-Felmayer G, Bogdan C. NKT cells mediate organ-specific resistance against Leishmania major infection. Microbes and Infection. 2006;8:354–362. doi: 10.1016/j.micinf.2005.07.002. [DOI] [PubMed] [Google Scholar]
- Mattner F, Magram J, Ferrante J, Launois P, Di Padova K, Behin R, et al. Genetically resistant mice lacking interleukin-12 are susceptible to infection with Leishmania major and mount a polarized Th2 cell response. European Journal of Immunology. 1996;26:1553–1559. doi: 10.1002/eji.1830260722. [DOI] [PubMed] [Google Scholar]
- Mbow ML, DeKrey GK, Titus RG. Leishmania major induces differential expression of costimulatory molecules on mouse epidermal cells. European Journal of Immunology. 2001;31:1400–1409. doi: 10.1002/1521-4141(200105)31:5<1400::aid-immu1400>3.0.co;2-j. [DOI] [PubMed] [Google Scholar]
- McDowell MA, Sacks DL. Inhibition of host cell signal transduction by Leishmania: observations relevant to the selective impairment of IL-12 responses. Current Opinion in Microbiology. 1999;2:438–443. doi: 10.1016/S1369-5274(99)80077-0. [DOI] [PubMed] [Google Scholar]
- Meddeb-Garnaoui A, Zrelli H, Dellagi K. Effects of tropism and virulence of Leishmania parasites on cytokine production by infected human monocytes. Clinical and Experimental Immunology. 2009;155:199–206. doi: 10.1111/j.1365-2249.2008.03821.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mendez S, Reckling SK, Piccirillo CA, Sacks D, Belkaid Y. Role for CD4(+) CD25(+) regulatory T cells in reactivation of persistent leishmaniasis and control of concomitant immunity. Journal of Experimental Medicine. 2004;200:201–210. doi: 10.1084/jem.20040298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mise-Omata S, Kuroda E, Sugiura T, Yamashita U, Obata Y, Doi TS. The NF-kappaB RelA subunit confers resistance to Leishmania major by inducing nitric oxide synthase 2 and Fas expression but not Th1 differentiation. Journal of Immunology. 2009;182:4910–4916. doi: 10.4049/jimmunol.0800967. [DOI] [PubMed] [Google Scholar]
- Mollinedo F, Janssen H, de la Iglesia-Vicente J, Villa-Pulgarin JA, Calafat J. Selective fusion of azurophilic granules with Leishmania-containing phagosomes in human neutrophils. Journal of Biological Chemistry. 2010;285:34528–34536. doi: 10.1074/jbc.M110.125302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moreno I, Molina R, Toraño A, Laurin E, García E, Domínguez M. Comparative real-time kinetic analysis of human complement killing of Leishmania infantum promastigotes derived from axenic culture or from Phlebotomus perniciosus. Microbes and Infection. 2007;9:1574–1580. doi: 10.1016/j.micinf.2007.09.009. [DOI] [PubMed] [Google Scholar]
- Mosser DM, Edelson PJ. The mouse macrophage receptor for C3bi (CR3) is a major mechanism in the phagocytosis of Leishmania promastigotes. Journal of Immunology. 1985;135:2785–2789. [PubMed] [Google Scholar]
- Müller K, van Zandbergen G, Hansen B, Laufs H, Jahnke N, Solbach W, et al. Chemokines, natural killer cells and granulocytes in the early course of Leishmania major infection in mice. Medical Microbiology and Immunology. 2001;190:73–76. doi: 10.1007/s004300100084. [DOI] [PubMed] [Google Scholar]
- Murphy ML, Cotterell SE, Gorak PM, Engwerda CR, Kaye PM. Blockade of CTLA-4 enhances host resistance to the intracellular pathogen, Leishmania donovani. Journal of Immunology. 1998;161:4153–4160. [PubMed] [Google Scholar]
- Murphy ML, Engwerda CR, Gorak PM, Kaye PM. B7-2 blockade enhances T cell responses to Leishmania donovani. Journal of Immunology. 1997;159:4460–4466. [PubMed] [Google Scholar]
- Murray HW, Nathan CF. Macrophage microbicidal mechanisms in vivo: reactive nitrogen versus oxygen intermediates in the killing of intracellular visceral Leishma-nia donovani. Journal of Experimental Medicine. 1999;189:741–746. doi: 10.1084/jem.189.4.741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nandan D, Lo R, Reiner NE. Activation of phosphotyrosine phosphatase activity attenuates mitogen-activated protein kinase signaling and inhibits c-FOS and nitric oxide synthase expression in macrophages infected with Leishmania donovani. Infection and Immunity. 1999;67:4055–4063. doi: 10.1128/iai.67.8.4055-4063.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nandan D, Reiner NE. Attenuation of gamma interferon-induced tyrosine phosphorylation in mononuclear phagocytes infected with Leishmania donovani: selective inhibition of signaling through Janus kinases and Stat1. Infection and Immunity. 1995;63:4495–4500. doi: 10.1128/iai.63.11.4495-4500.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nateghi Rostami M, Keshavarz H, Edalat R, Sarrafnejad A, Shahrestani T, Mahboudi F, et al. CD8+ T cells as a source of IFN-γ production in human cutaneous leishmaniasis. PLoS Neglected Tropical Diseases. 2010;4:e845. doi: 10.1371/journal.pntd.0000845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Novais FO, Santiago RC, Báfica A, Khouri R, Afonso L, Borges VM, et al. Neutrophils and macrophages cooperate in host resistance against Leishmania braziliensis infection. Journal of Immunology. 2009;183:8088–8098. doi: 10.4049/jimmunol.0803720. [DOI] [PubMed] [Google Scholar]
- Oghumu S, Lezama-Dávila CM, Isaac-Márquez AP, Satoskar AR. Role of chemokines in regulation of immunity against leishmaniasis. Experimental Parasitology. 2010;126:389–396. doi: 10.1016/j.exppara.2010.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olivier M, Atayde VD, Isnard A, Hassani K, Shio MT. Leishmania virulence factors: focus on the metalloprotease GP63. Microbes and Infection. 2012;14:1377–1389. doi: 10.1016/j.micinf.2012.05.014. [DOI] [PubMed] [Google Scholar]
- Olivier M, Brownsey RW, Reiner NE. Defective stimulus-response coupling in human monocytes infected with Leishmania donovani is associated with altered activation and translocation of protein kinase C. Proceedings of the National Academy of Sciences of the United States of America. 1992;89:7481–7485. doi: 10.1073/pnas.89.16.7481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olivier M, Gregory DJ, Forget G. Subversion mechanisms by which Leishmania parasites can escape the host immune response: a signaling point of view. Clinical Microbiology Reviews. 2005;18:293–305. doi: 10.1128/CMR.18.2.293-305.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pearson RD, Steigbigel RT. Phagocytosis and killing of the protozoan Leishmania donovani by human polymorphonuclear leukocytes. Journal of Immunology. 1981;127:1438–1443. [PubMed] [Google Scholar]
- Pereira LI, Dorta ML, Pereira AJ, Bastos RP, Oliveira MA, Pinto SA, et al. Increase of NK cells and proinflammatory monocytes are associated with the clinical improvement of diffuse cutaneous leishmaniasis after immunochemotherapy with BCG/Leishmania antigens. American Journal of Tropical Medicine and Hygiene. 2009;81:378–383. [PubMed] [Google Scholar]
- Pereira WF, Ribeiro-Gomes FL, Guillermo LV, Vellozo NS, Montalvão F, Dosreis GA, et al. Myeloid-derived suppressor cells help protective immunity to Leishmania major infection despite suppressed T cell responses. Journal of Leukocyte Biology. 2011;90:1191–1197. doi: 10.1189/jlb.1110608. [DOI] [PubMed] [Google Scholar]
- Peruhype-Magalhães V, Martins-Filho OA, Prata A, Silva LeA, Rabello A, Teixeira-Carvalho A, et al. Immune response in human visceral leishmaniasis: analysis of the correlation between innate immunity cytokine profile and disease outcome. Scandinavian Journal of Immunology. 2005;62:487–495. doi: 10.1111/j.1365-3083.2005.01686.x. [DOI] [PubMed] [Google Scholar]
- Peters N, Sacks D. Immune privilege in sites of chronic infection: leishmania and regulatory T cells. Immunological Reviews. 2006;213:159–179. doi: 10.1111/j.1600-065X.2006.00432.x. [DOI] [PubMed] [Google Scholar]
- Pinheiro NF, Hermida MD, Macedo MP, Mengel J, Bafica A, dos-Santos WL. Leishmania infection impairs beta 1-integrin function and chemokine receptor expression in mononuclear phagocytes. Infection and Immunity. 2006;74:3912–3921. doi: 10.1128/IAI.02103-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pompeu ML, Freitas LA, Santos ML, Khouri M, Barral-Netto M. Granulocytes in the inflammatory process of BALB/c mice infected by Leishmania amazonensis. A quantitative approach. Acta Tropica. 1991;48:185–193. doi: 10.1016/0001-706x(91)90046-m. [DOI] [PubMed] [Google Scholar]
- Prajeeth CK, Haeberlein S, Sebald H, Schleicher U, Bogdan C. Leishmania-infected macrophages are targets of NK cell-derived cytokines but not of NK cell cytotoxicity. Infection and Immunity. 2011;79:2699–2708. doi: 10.1128/IAI.00079-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prina E, Lang T, Glaichenhaus N, Antoine JC. Presentation of the protective parasite antigen LACK by Leishmania-infected macrophages. Journal of Immunology. 1996;156:4318–4327. [PubMed] [Google Scholar]
- Privé C, Descoteaux A. Leishmania donovani promastigotes evade the activation of mitogen-activated protein kinases p38, c-Jun N-terminal kinase, and extracellular signal-regulated kinase-1/2 during infection of naive macrophages. European Journal of Immunology. 2000;30:2235–2244. doi: 10.1002/1521-4141(2000)30:8<2235::AID-IMMU2235>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
- Puentes SM, Da Silva RP, Sacks DL, Hammer CH, Joiner KA. Serum resistance of metacyclic stage Leishmania major promastigotes is due to release of C5b-9. Journal of Immunology. 1990;145:4311–4316. [PubMed] [Google Scholar]
- Puentes SM, Sacks DL, da Silva RP, Joiner KA. Complement binding by two developmental stages of Leishmania major promastigotes varying in expression of a surface lipophosphoglycan. Journal of Experimental Medicine. 1988;167:887–902. doi: 10.1084/jem.167.3.887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rai AK, Thakur CP, Seth T, Mitra DK. Enrichment of invariant natural killer T cells in the bone marrow of visceral leishmaniasis patients. Parasite Immunology. 2011;33:688–691. doi: 10.1111/j.1365-3024.2011.01328.x. [DOI] [PubMed] [Google Scholar]
- Rai AK, Thakur CP, Singh A, Seth T, Srivastava SK, Singh P, et al. Regulatory T cells suppress T cell activation at the pathologic site of human visceral leishmaniasis. PLoS One. 2012;7:e31551. doi: 10.1371/journal.pone.0031551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raman VS, Bhatia A, Picone A, Whittle J, Bailor HR, O’Donnell J, et al. Applying TLR synergy in immunotherapy: implications in cutaneous leishmaniasis. Journal of Immunology. 2010;185:1701–1710. doi: 10.4049/jimmunol.1000238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reguera RM, Balaña-Fouce R, Showalter M, Hickerson S, Beverley SM. Leishmania major lacking arginase (ARG) are auxotrophic for polyamines but retain infectivity to susceptible BALB/c mice. Molecular and Biochemical Parasitology. 2009;165:48–56. doi: 10.1016/j.molbiopara.2009.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reiner SL, Zheng S, Wang ZE, Stowring L, Locksley RM. Leishmania promastigotes evade interleukin 12 (IL-12) induction by macrophages and stimulate a broad range of cytokines from CD4+ T cells during initiation of infection. Journal of Experimental Medicine. 1994;179:447–456. doi: 10.1084/jem.179.2.447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rezai HR, Sher S, Gettner S. Leishmania tropica, L. donovani, and L. enriettii: immune rabbit serum inhibitory in vitro. Experimental Parasitology. 1969;26:257–263. doi: 10.1016/0014-4894(69)90117-9. [DOI] [PubMed] [Google Scholar]
- Ribeiro-Gomes FL, Moniz-de-Souza MC, Alexandre-Moreira MS, Dias WB, Lopes MF, Nunes MP, et al. Neutrophils activate macrophages for intracellular killing of Leishmania major through recruitment of TLR4 by neutrophil elastase. Journal of Immunology. 2007;179:3988–3994. doi: 10.4049/jimmunol.179.6.3988. [DOI] [PubMed] [Google Scholar]
- 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. Journal of Immunology. 2004;172:4454–4462. doi: 10.4049/jimmunol.172.7.4454. [DOI] [PubMed] [Google Scholar]
- Ritter U, Moll H, Laskay T, Bröcker E, Velazco O, Becker I, et al. Differential expression of chemokines in patients with localized and diffuse cutaneous American leishmaniasis. Journal of Infectious Diseases. 1996;173:699–709. doi: 10.1093/infdis/173.3.699. [DOI] [PubMed] [Google Scholar]
- Sacks DL, Pimenta PF, McConville MJ, Schneider P, Turco SJ. Stage-specific binding of Leishmania donovani to the sand fly vector midgut is regulated by conformational changes in the abundant surface lipophosphoglycan. Journal of Experimental Medicine. 1995;181:685–697. doi: 10.1084/jem.181.2.685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saha B, Das G, Vohra H, Ganguly NK, Mishra GC. Macrophage-T cell interaction in experimental visceral leishmaniasis: failure to express costimulatory molecules on Leishmania-infected macrophages and its implication in the suppression of cell-mediated immunity. European Journal of Immunology. 1995;25:2492–2498. doi: 10.1002/eji.1830250913. [DOI] [PubMed] [Google Scholar]
- Satoskar AR, Stamm LM, Zhang X, Okano M, David JR, Terhorst C, et al. NK cell-deficient mice develop a Th1-like response but fail to mount an efficient antigen-specific IgG2a antibody response. Journal of Immunology. 1999;163:5298–5302. [PubMed] [Google Scholar]
- Scharton TM, Scott P. Natural killer cells are a source of interferon gamma that drives differentiation of CD4+ T cell subsets and induces early resistance to Leishmania major in mice. Journal of Experimental Medicine. 1993;178:567–577. doi: 10.1084/jem.178.2.567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scianimanico S, Desrosiers M, Dermine JF, Méresse S, Descoteaux A, Desjardins M. Impaired recruitment of the small GTPase rab7 correlates with the inhibition of phagosome maturation by Leishmania donovani promastigotes. Cellular Microbiology. 1999;1:19–32. doi: 10.1046/j.1462-5822.1999.00002.x. [DOI] [PubMed] [Google Scholar]
- Sen S, Roy K, Mukherjee S, Mukhopadhyay R, Roy S. Restoration of IFNγR subunit assembly, IFNγ signaling and parasite clearance in Leishmania donovani infected macrophages: role of membrane cholesterol. PLoS Pathogens. 2011;7:e1002229. doi: 10.1371/journal.ppat.1002229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shakya N, Sane SA, Shankar S, Gupta S. Effect of Pam3Cys induced protection on the therapeutic efficacy of miltefosine against experimental visceral leishmaniasis. Peptides. 2011;32:2131–2133. doi: 10.1016/j.peptides.2011.09.014. [DOI] [PubMed] [Google Scholar]
- Shweash M, Adrienne McGachy H, Schroeder J, Neamatallah T, Bryant CE, Millington O, et al. Leishmania mexicana promastigotes inhibit macrophage IL-12 production via TLR-4 dependent COX-2, iNOS and arginase-1 expression. Molecular Immunology. 2011;48:1800–1808. doi: 10.1016/j.molimm.2011.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silva VM, Larangeira DF, Oliveira PR, Sampaio RB, Suzart P, Nihei JS, et al. Enhancement of experimental cutaneous leishmaniasis by Leishmania molecules is dependent on interleukin-4, serine protease/esterase activity, and parasite and host genetic backgrounds. Infection and Immunity. 2011;79:1236–1243. doi: 10.1128/IAI.00309-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smelt SC, Cotterell SE, Engwerda CR, Kaye PM. B cell-deficient mice are highly resistant to Leishmania donovani infection, but develop neutrophil-mediated tissue pathology. Journal of Immunology. 2000;164:3681–3688. doi: 10.4049/jimmunol.164.7.3681. [DOI] [PubMed] [Google Scholar]
- de Souza Carmo EV, Katz S, Barbiéri CL. Neutrophils reduce the parasite burden in Leishmania (Leishmania) amazonensis-infected macrophages. PLoS One. 2010;5:e13815. doi: 10.1371/journal.pone.0013815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srivastava A, Singh N, Mishra M, Kumar V, Gour JK, Bajpai S, et al. Identification of TLR inducing Th1-responsive Leishmania donovani amastigote-specific antigens. Molecular and Cellular Biochemistry. 2012;359:359–368. doi: 10.1007/s11010-011-1029-5. [DOI] [PubMed] [Google Scholar]
- Srivastava N, Sudan R, Saha B. CD40-modulated dual-specificity phosphatases MAPK phosphatase (MKP)-1 and MKP-3 reciprocally regulate Leishmania major infection. Journal of Immunology. 2011;186:5863–5872. doi: 10.4049/jimmunol.1003957. [DOI] [PubMed] [Google Scholar]
- Srivastav S, Kar S, Chande AG, Mukhopadhyaya R, Das PK. Leishmania donovani exploits host deubiquitinating enzyme A20, a negative regulator of TLR signaling, to subvert host immune response. Journal of Immunology. 2012;189:924–934. doi: 10.4049/jimmunol.1102845. [DOI] [PubMed] [Google Scholar]
- Stanley AC, Zhou Y, Amante FH, Randall LM, Haque A, Pellicci DG, et al. Activation of invariant NKT cells exacerbates experimental visceral leishmaniasis. PLoS Pathogens. 2008;4:e1000028. doi: 10.1371/journal.ppat.1000028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suffia I, Reckling SK, Salay G, Belkaid Y. A role for CD103 in the retention of CD4+CD25+ Treg and control of Leishmania major infection. Journal of Immunology. 2005;174:5444–5455. doi: 10.4049/jimmunol.174.9.5444. [DOI] [PubMed] [Google Scholar]
- Suzuki E, Tanaka AK, Toledo MS, Takahashi HK, Straus AH. Role of beta-D-galactofuranose in Leishmania major macrophage invasion. Infection and Immunity. 2002;70:6592–6596. doi: 10.1128/IAI.70.12.6592-6596.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swihart K, Fruth U, Messmer N, Hug K, Behin R, Huang S, et al. Mice from a genetically resistant background lacking the interferon gamma receptor are susceptible to infection with Leishmania major but mount a polarized T helper cell 1-type CD4+ T cell response. Journal of Experimental Medicine. 1995;181:961–971. doi: 10.1084/jem.181.3.961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tabatabaee PA, Abolhassani M, Mahdavi M, Nahrevanian H, Azadmanesh K. Leishmania major: secreted antigens of Leishmania major promastigotes shift the immune response of the C57BL/6 mice toward Th2 in vitro. Experimental Parasitology. 2011;127:46–51. doi: 10.1016/j.exppara.2010.06.033. [DOI] [PubMed] [Google Scholar]
- Tacchini-Cottier F, Zweifel C, Belkaid Y, Mukankundiye C, Vasei M, Launois P, et al. An immunomodulatory function for neutrophils during the induction of a CD4+ Th2 response in BALB/c mice infected with Leishmania major. Journal of Immunology. 2000;165:2628–2636. doi: 10.4049/jimmunol.165.5.2628. [DOI] [PubMed] [Google Scholar]
- Talamás-Rohana P, Wright SD, Lennartz MR, Russell DG. Lipophosphoglycan from Leishmania mexicana promastigotes binds to members of the CR3, p150,95 and LFA-1 family of leukocyte integrins. Journal of Immunology. 1990;144:4817–4824. [PubMed] [Google Scholar]
- Thalhofer CJ, Chen Y, Sudan B, Love-Homan L, Wilson ME. Leukocytes infiltrate the skin and draining lymph nodes in response to the protozoan Leishmania infantum chagasi. Infection and Immunity. 2011;79:108–117. doi: 10.1128/IAI.00338-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsagozis P, Karagouni E, Dotsika E. Function of CD8+ T lymphocytes in a self-curing mouse model of visceral leishmaniasis. Parasitology International. 2005;54:139–146. doi: 10.1016/j.parint.2005.02.005. [DOI] [PubMed] [Google Scholar]
- Turco SJ, Hull SR, Orlandi PA, Shepherd SD, Homans SW, Dwek RA, et al. Structure of the major carbohydrate fragment of the Leishmania donovani lipophosphoglycan. Biochemistry. 1987;26:6233–6238. doi: 10.1021/bi00393a042. [DOI] [PubMed] [Google Scholar]
- Vargas-Inchaustegui DA, 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. Infection and Immunity. 2009;77:2948–2956. doi: 10.1128/IAI.00154-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Veer MJ, Curtis JM, Baldwin TM, DiDonato JA, Sexton A, McConville MJ, et al. MyD88 is essential for clearance of Leishmania major: possible role for lipophosphoglycan and toll-like receptor 2 signaling. European Journal of Immunology. 2003;33:2822–2831. doi: 10.1002/eji.200324128. [DOI] [PubMed] [Google Scholar]
- Vinet AF, Fukuda M, Turco SJ, Descoteaux A. The Leishmania donovani lipophosphoglycan excludes the vesicular proton-ATPase from phagosomes by impairing the recruitment of synaptotagmin V. PLoS Pathogens. 2009;5:e1000628. doi: 10.1371/journal.ppat.1000628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vivarini AeC, Pereira ReM, Teixeira KL, Calegari-Silva TC, Bellio M, Laurenti MD, et al. Human cutaneous leishmaniasis: interferon-dependent expression of double-stranded RNA-dependent protein kinase (PKR) via TLR2. FASEB Journal. 2011;25:4162–4173. doi: 10.1096/fj.11-185165. [DOI] [PubMed] [Google Scholar]
- Weingartner A, Kemmer G, Muller FD, Zampieri RA, Gonzaga Dos Santos M, Schiller J, et al. Leishmania promastigotes lack phosphatidylserine but bind annexin V upon permeabilization or miltefosine treatment. PLoS One. 2012;7:e42070. doi: 10.1371/journal.pone.0042070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weinheber N, Wolfram M, Harbecke D, Aebischer T. Phagocytosis of Leishmania mexicana amastigotes by macrophages leads to a sustained suppression of IL-12 production. European Journal of Immunology. 1998;28:2467–2477. doi: 10.1002/(SICI)1521-4141(199808)28:08<2467::AID-IMMU2467>3.0.CO;2-1. [DOI] [PubMed] [Google Scholar]
- Whitaker SM, Colmenares M, Pestana KG, McMahon-Pratt D. Leishmania pifanoi proteoglycolipid complex P8 induces macrophage cytokine production through toll-like receptor 4. Infection and Immunity. 2008;76:2149–2156. doi: 10.1128/IAI.01528-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wiethe C, Debus A, Mohrs M, Steinkasserer A, Lutz M, Gessner A. Dendritic cell differentiation state and their interaction with NKT cells determine Th1/Th2 differentiation in the murine model of Leishmania major infection. Journal of Immunology. 2008;180:4371–4381. doi: 10.4049/jimmunol.180.7.4371. [DOI] [PubMed] [Google Scholar]
- Wilson J, Huynh C, Kennedy KA, Ward DM, Kaplan J, Aderem A, et al. Control of parasitophorous vacuole expansion by LYST/Beige restricts the intracellular growth of Leishmania amazonensis. PLoS Pathogens. 2008;4:e1000179. doi: 10.1371/journal.ppat.1000179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wright SD, Silverstein SC. Receptors for C3b and C3bi promote phagocytosis but not the release of toxic oxygen from human phagocytes. Journal of Experimental Medicine. 1983;158:2016–2023. doi: 10.1084/jem.158.6.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Z, Mosser DM, Zhang X. Activation of the MAPK, ERK, following Leishmania amazonensis infection of macrophages. Journal of Immunology. 2007;178:1077–1085. doi: 10.4049/jimmunol.178.2.1077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yurchenko E, Tritt M, Hay V, Shevach EM, Belkaid Y, Piccirillo CA. CCR5-dependent homing of naturally occurring CD4+ regulatory T cells to sites of Leishmania major infection favors pathogen persistence. Journal of Experimental Medicine. 2006;203:2451–2460. doi: 10.1084/jem.20060956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Zandbergen G, Hermann N, Laufs H, Solbach W, Laskay T. Leishmania promastigotes release a granulocyte chemotactic factor and induce interleukin-8 release but inhibit gamma interferon-inducible protein 10 production by neutrophil granulocytes. Infection and Immunity. 2002;70:4177–4184. doi: 10.1128/IAI.70.8.4177-4184.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]