role of exogenous chemokines as immunotherapeutic tool ... · role of exogenous chemokines as...

8
Role of exogenous chemokines as immunotherapeutic tool against visceral leishmaniasis. G. Gupta 1 , R. Dey 2 , S. Bhattacharyya 3 and S. Majumdar 1* 1 Division of Molecular Medicine, Centenary Campus, Bose Institute, Kolkata, India. 2 Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, FDA, Bethesda, MD, USA 3 Department of Pediatrics, Washington University, School of Medicine, St. Louis, Missori, USA. Visceral leishmaniasis (VL), caused by the protozoan parasite, Leishmania donovani, is associated with immunological dysfunctions. Recent observations in humans and mice suggest a rapid change in chemokine expression, pointing towards their possible involvement in the disease process. Chemokines form an integral part of the host defense against pathogens and the release of chemokines is regarded as a crucial step in cellular recruitment thereby facilitatiting the initiation and maintenance of inflammatory responses that is required for counteracting the microbial invaders. Here in this review, we have highlighted the importance of the exogenous CC and CXC chemokines as potential immunotherapeutic tools to counteract Leishmania induced pathogenesis. Key words: Leishmania; chemokines; macrophages; immune response. 1. Visceral leishmaniasis (VL): 1.1 Overview: Leishmaniasis, a tropical disease, is one of the major health problems facing mankind which occurs in the skin as localized cutaneous (CL) and mucocutaneous (MCL) leishmaniasis to diffuse cutaneous leishmaniasis (DCL), whereas in the viscera they range from subclinical to potentially fatal disease [1]. World Health Organization estimates an incidence of 12 million cases among 350 million at risk and an annual incidence of 1.5 million to 2 million new cases of CL and 500,000 new cases of VL [2]. Leishmaniasis is caused by protozoan parasites belonging to the genus Leishmania. These parasites are transmitted by the bite of a Phlebotomine sandfly [3]. VL, the most severe form of leishmaniasis, is caused by Leishmania donovani and Leishmania chagasi in the Old and New worlds, respectively [4, 5] and is associated with high parasite numbers and the absence of an effective T helper cell type 1 (Th1) immune response [1] 2. Intracellular survival of Leishmania parasites: Leishmania has the ability to survive in the macrophageal lysosomal compartment, an intracellular site normally inhospitable to invading microorganisms [4]. Intracellular parasitism of macrophages by Leishmania culminates in all the clinical and pathological symptoms associated with leishmaniasis and is favored by several parasites derived factors [6-8]. The inflammatory reactions initiated by the host macrophage during the entry of Leishmania occur in two stages. Firstly, during the initial uptake and phagocytosis of promastigotes, the macrophage produces toxic free radicals, including superoxide anions (O 2 - ) [9, 10]. Exposure to O 2 - has been reported to be fatal to the Leishmania promastigotes [9-11] but its generation is inhibited by L. donovani infection [12, 13] which appears to be dependent upon cell surface molecules, lipophosphoglycan (LPG) and glycoprotein (GP) 63 [14, 15]. Secondly, even after infection is established, the quiescent macrophage can be activated to kill intracellular amastigote form of L. donovani. This second anti- leishmanial event occurs via the NO generation after activation of macrophages by CC chemokines, interferon (IFN)- or tumor necrosis factor (TNF)-α along with lipopolysaccharide (LPS) [16-19]. Infected macrophages incubated with purified Leishmania surface molecules, LPG or glycosylphosphoionsitol (GIPL) lose their ability to activate inducible nitric oxide synthase (iNOS) [20, 21]. 2.1 Cytokine and chemokine mediated regulation of disease: During Leishmania infection, cytokines regulate the host response. The important inflammatory cytokines released by Leishmania infected macrophages include interleukin (IL)-1β, TNF-α and IL-12. Additionally, Th1 cells produce IFN- whereas; Th2 cells produce IL-4. Dendritic cell derived IL-12 by and natural killer (NK) cell derived IFN- are also critical in determining the outcome of the disease [22, 23]. Cytokines have a direct involvement in chemokine production and can also precede the expression of some chemokines, which in turn, induces the production of additional 605 ©FORMATEX 2011 Science against microbial pathogens: communicating current research and technological advances A. Méndez-Vilas (Ed.) _______________________________________________________________________________

Upload: truongthuan

Post on 13-Feb-2019

218 views

Category:

Documents


0 download

TRANSCRIPT

Role of exogenous chemokines as immunotherapeutic tool against visceral leishmaniasis.

G. Gupta1 , R. Dey2, S. Bhattacharyya3 and S. Majumdar1*

1 Division of Molecular Medicine, Centenary Campus, Bose Institute, Kolkata, India. 2 Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, FDA, Bethesda, MD, USA 3 Department of Pediatrics, Washington University, School of Medicine, St. Louis, Missori, USA.

Visceral leishmaniasis (VL), caused by the protozoan parasite, Leishmania donovani, is associated with immunological dysfunctions. Recent observations in humans and mice suggest a rapid change in chemokine expression, pointing towards their possible involvement in the disease process. Chemokines form an integral part of the host defense against pathogens and the release of chemokines is regarded as a crucial step in cellular recruitment thereby facilitatiting the initiation and maintenance of inflammatory responses that is required for counteracting the microbial invaders. Here in this review, we have highlighted the importance of the exogenous CC and CXC chemokines as potential immunotherapeutic tools to counteract Leishmania induced pathogenesis.

Key words: Leishmania; chemokines; macrophages; immune response.

1. Visceral leishmaniasis (VL):

1.1 Overview:

Leishmaniasis, a tropical disease, is one of the major health problems facing mankind which occurs in the skin as localized cutaneous (CL) and mucocutaneous (MCL) leishmaniasis to diffuse cutaneous leishmaniasis (DCL), whereas in the viscera they range from subclinical to potentially fatal disease [1]. World Health Organization estimates an incidence of 12 million cases among 350 million at risk and an annual incidence of 1.5 million to 2 million new cases of CL and 500,000 new cases of VL [2]. Leishmaniasis is caused by protozoan parasites belonging to the genus Leishmania. These parasites are transmitted by the bite of a Phlebotomine sandfly [3]. VL, the most severe form of leishmaniasis, is caused by Leishmania donovani and Leishmania chagasi in the Old and New worlds, respectively [4, 5] and is associated with high parasite numbers and the absence of an effective T helper cell type 1 (Th1) immune response [1]

2. Intracellular survival of Leishmania parasites:

Leishmania has the ability to survive in the macrophageal lysosomal compartment, an intracellular site normally inhospitable to invading microorganisms [4]. Intracellular parasitism of macrophages by Leishmania culminates in all the clinical and pathological symptoms associated with leishmaniasis and is favored by several parasites derived factors [6-8]. The inflammatory reactions initiated by the host macrophage during the entry of Leishmania occur in two stages. Firstly, during the initial uptake and phagocytosis of promastigotes, the macrophage produces toxic free radicals, including superoxide anions (O2

-) [9, 10]. Exposure to O2- has been reported to be fatal to the Leishmania promastigotes

[9-11] but its generation is inhibited by L. donovani infection [12, 13] which appears to be dependent upon cell surface molecules, lipophosphoglycan (LPG) and glycoprotein (GP) 63 [14, 15]. Secondly, even after infection is established, the quiescent macrophage can be activated to kill intracellular amastigote form of L. donovani. This second anti-leishmanial event occurs via the NO generation after activation of macrophages by CC chemokines, interferon (IFN)- or tumor necrosis factor (TNF)-α along with lipopolysaccharide (LPS) [16-19]. Infected macrophages incubated with purified Leishmania surface molecules, LPG or glycosylphosphoionsitol (GIPL) lose their ability to activate inducible nitric oxide synthase (iNOS) [20, 21].

2.1 Cytokine and chemokine mediated regulation of disease:

During Leishmania infection, cytokines regulate the host response. The important inflammatory cytokines released by Leishmania infected macrophages include interleukin (IL)-1β, TNF-α and IL-12. Additionally, Th1 cells produce IFN- whereas; Th2 cells produce IL-4. Dendritic cell derived IL-12 by and natural killer (NK) cell derived IFN- are also critical in determining the outcome of the disease [22, 23]. Cytokines have a direct involvement in chemokine production and can also precede the expression of some chemokines, which in turn, induces the production of additional

605©FORMATEX 2011

Science against microbial pathogens: communicating current research and technological advances A. Méndez-Vilas (Ed.)_______________________________________________________________________________

inflammatory mediators. Cytokines exert a secondary effect on leukocyte recruitment by inducing the expression of several chemokine genes [24]. In leishmaniasis, cytokines seem to synergize with leishmanial elements to regulate chemokine production. TNF-α and IL-1β, together with macrophage inflammatory protein (MIP)-1α, regulate Langerhans cell-mediated transport of Leishmania from the infected skin to regional lymph nodes (LNs) in murine CL [25]. IL-12 is required for the induction of Th1-related chemokines such as lymphotactin, IFN--inducible protein 10 (CXCL10 or IP-10) and monocyte chemotactic protein 1 (MCP-1) in LNs of resistant L. major-infected mice [26]. Interestingly, Th1- and Th2-derived cytokines can have antagonistic effects on chemokines. Monokine induced by IFN- (MIG) and CXCL10 are more selectively induced by IFN- [27]. The Th2-related cytokines IL-4 and IL-13 induce macrophage-derived chemokine (MDC) and CCL6 production in macrophages, and this production is inhibited by IFN- [28, 29].

2.1.1 Modulation of host cell signaling:

Several signaling molecules have been implicated in the regulation of antiparasitic host immune response, including members of the protein kinase C (PKC) super-family and mitogen activated protein kinase (MAPK) family [30-33]. MAPKs are a group of serine/threonine kinases, the enzymatic activity of which is elicited upon phosphorylation of threonine and tyrosine residues in a Thr-X-Tyr motif in their regulatory domain [34]. It includes extracellular signal-related kinase 1 and 2 (ERK1/2), c-jun NH2-terminal kinase (JNK) and p38MAPK. MAPK phosphorylate selected intracellular proteins, including transcription factors, which subsequently regulate gene expression by transcriptional and post-transcriptional mechanisms [35]. MAPKs are actively repressed and cannot be activated when Leishmania-infected macrophages are stimulated with a variety of agonists. Inhibition of MAPK phosphorylation resulted in low expression of IL-12 and iNOS2 [36, 37] which has been shown to play crucial role in the development of immunity to Leishmania major [38]. CD40-induced p38MAPK phosphorylation, iNOS2 expression, and antileishmanial function were impaired in Leishmania major-infected macrophages but were restored by anisomycin, a p38MAPK activator, suggesting a crucial role of p38MAPK in CD40 signaling. Anisomycin’s effects were reversed by SB203580, a p38MAPK-specific inhibitor, emphasizing the role of p38MAPK in CD40-induced iNOS2-dependent leishmanicidal function [39]. Activation of MAPKs depends on PKC [35], suggesting that Leishmania parasites can impair signaling through these enzymes in infected cells. PKC is a calcium and phospholipid dependent serine / threonine kinase that exists as a family of different isotypes having closely related structure (30, 32, 40). The fourteen known isoforms of PKC family have been subdivided into the classical (cPKC; α, β-I, β-II, ), novel (nPKC; , , , , ) and atypical (aPKC; , , ) groups. Infection with L. donovani or Leishmania derived glycolipid, lipophosphoglycan (LPG) accounted for impaired PKC activity [41, 42]. PKC regulates phagosome maturation, as the iso-enzymes α-and β-PKC are associated with phagosomal membrane [43, 44]. Leishmania and Leishmania derived LPG impairs the PKC dependent signal transduction in macrophages thus helping the parasite to survive in the macrophageal microenvironment [12-14, 31]. Accordingly, BALB/c peritoneal macrophages infected with UR-6, LPG deficient attenuated leishmanial parasite [42, 45], enhanced the PKC-β activity. The decrease in Ca2+-dependent PKC β activity might be due to IL-10 produced by L. donovani infection as pretreatment with anti-IL-10 neutralizing antibody significantly restored Ca2+-dependent PKC β activity [45].

3. Chemokines:

3.1 Classification and structure:

Chemokines are a homologous super family of relatively small proteins ranging from 8 to 17 kDa [46, 47]. The super family of chemokines is subclassified on the basis of the arrangement of cysteine residues located in the N-terminal regions of these molecules. These are designated C, CC, CXC, and CXXC, where C represents the number of N-terminal region cysteine residues and X represents the number of intervening amino acids [47-49]. CXC (α) chemokines have their first two consensus cysteines separated by an amino acid; the CC (β) chemokines have their first two cysteines adjacent to each other. The other two minor subfamilies include the CX3C chemokines and the C chemokines [50]. Chemokines exert their effect through guanosine nucleotide-protein coupled receptors (GPCR) [48]. Like all known GPCR, the chemokine receptors have seven transmembrane hydrophobic domains with three intracellular and three extracellular hydrophilic loops.

606 ©FORMATEX 2011

Science against microbial pathogens: communicating current research and technological advances A. Méndez-Vilas (Ed.)______________________________________________________________________________

Table 1. Homologue of human and mouse chemokines

3.1.1 General functions:

Chemokines regulate distinct signal transduction pathways culminating in a variety of biological functions including integrin activation and cell migration along a chemokine gradient [51]. They provide the directional cues for the movement of leukocytes in development, homeostatis, and inflammation. Leukocytes extravasation from the blood into the tissues is a regulated multistep process involving a series of coordinated interactions between leukocytes and endothelial cells [49, 52]. Extravasated leukocytes follow a gradient of chemokine concentration and come to rest at a site of high concentration [52]. The dramatic increase in the secretion of chemokines during inflammation results in the selective recruitment of leukocytes into inflamed tissue [48, 53]. They play a vital role in the regulation of immunity to various diseases [54-56] where distinct chemokines and chemokine receptor-integrin combinations are associated with particular diseases by controlling the effector cell migration to their respective infected tissue sites [50].

Human Mouse Homologue Systematic Name

C-X-C group or α group

Growth–related oncogenes (GRO) α (MGSA),

β (MIP-2α), and g (MIP-2β)

Platelet factor 4 (PF-4)

Granulocyte chemotactic protein (GCP-2)

Neutrophil-activating protein (NAP-2)

Interleukin-8 (IL-8)

Monokine induced by IFN-γ (MIG)

IFN-γ-inducible protein (IP-10)

IFN-γ-induced Tcell alpha chemokine (I-TAC)

Stromal cell-derived factor (SDF-1)

mKC, mMIP-2

mPF-4

mGCP

Unknown

Unknown

mMIG

mIP-10 (mCRG-2)

Unknown

mSDF-1

CXCL1, CXCL2

CXCL4

CXCL6

CXCL7

CXCL8

CXCL9

CXCL10

CXCL11

CXCL12

C-C group or β group

Monocyte chemotactic protein-1 (MCP-1)

Macrophage inflammatory protein 1α (MIP-1α)

Macrophage inflammatory protein 1 β (MIP-1β)

Regulated on activation and normally T-cell expressed

(RANTES)

Macrophage inflammatory protein 1d (MIP-1d)

Monocyte chemotactic protein 3 (MCP-3)

Monocyte chemotactic protein 2 (MCP-2)

Monocyte chemotactic protein 4 (MCP-4)

Hepatocyte chemokines (HCC-1,HCC-2)

HCC-4

Thymus and activation related (TARC)

Pulmonary and activation related (PARC)

Monocyte derived (MDC)

Eotoxin-3

Thymus-expressed chemokine (TECK)

mMCP-1/JE

mMIP -1 α

mMIP -1β

mRANTES

mC10

mMCP-3

mMCP-2

Unknown

Unknown

mTARC

Unknown

mABCD-1

unknown

mTECK

CCL2

CCL3

CCL4

CCL5

CCL6

CCL7

CCL8

CCL13

CCL14,15

CCL16

CCL17

CCL18

CCL22

CCL25

CCL26

C group or γ group

Lymphotactin

mLymphotactin

XCL1

C-X-X-C or δ group

Fractalkine

mFractalkine

CX3CL1

607©FORMATEX 2011

Science against microbial pathogens: communicating current research and technological advances A. Méndez-Vilas (Ed.)_______________________________________________________________________________

Fig .1 Biological functions of chemokines

3.1.2 Protective role in different diseases:

Chemokines form an integral part of the host defense against pathogenic invasion. In bacterial and fungal infections, a rapid inflammatory response is required to counteract the microbial invaders. Under such a situation an induction of chemokine has been observed in several animal models as well as clinical studies [57-59]. Chemokines were found to be induced in the lungs of patients with Mycobacterium tuberculosis infection, in which they are secreted by bronchoalveolar macrophage [59]. Recent studies using chemokine have been focused on blockade or use recombinant chemokine treatment for infectious disease. MCP-1, a CC chemokine, restored the impaired free radical generation and proinflammatory cytokines in mycobacterial pathogenesis [60]. MCP-1 also regulates the pulmonary host defense via neutrophil recruitment during E. coli infection [61]. CC chemokines contribute to the formation of mononuclear phagocyte dependent granulomas viz. MIP-1α is found to induce granuloma in mouse infected with Schistosoma mansoni [62]. In MIP-1α knockout mice a pronounced delay in resolution of viral infection was observed, which were associated with substantially reduced recruitment of CD8+ T cells into the infected lung [63]. Studies have suggested a potentially important role of CC chemokines, MIP-1α, MIP-1β, and RANTES have been identified as suppressive factors for HIV-1 by inhibiting the replication of the macrophage tropic HIV strains [64]. Moreover MIP-1α, MIP-1β and RANTES activate human macrophages to kill Trypanosoma cruzi via NO dependent mechanism [65]. Co-injecting CXCL10 with Vaccinia antigens activates the cytotoxic T cells which render protection against subsequent infectious challenge with vaccinia virus [66].

4. Chemokines and Visceral leishmaniasis

4.1 Changes in chemokine expression during VL:

Initial phase of L. donovani infection is marked by an induction of MIP-1α and MCP-1 which attracts the monocytic cells as a source of Th1 mobilizing chemokines such as CXCL10 [67]. This is accompanied by CCR7 downregulation leading to the impairment of DC migration to lymph nodes contributing to disease progression [68]. Moreover in CCR5-

/-, MIP-1α-/- or CCR2-/- knockout mice there was low antigen specific IFN-γ response during early L. donovani infection [69]. During the late phase, L. donovani infected mice undergo a rapid hepatic accumulation of MIP-1α, MCP-1 and CXCL10 [67, 69]. Whereas, VL patients show elevated concentrations of CXCL9 and CXCL10 in their serum during active infection L. donovani which is critical for pathogenesis [70], thereby suggesting differences in chemokine expression in experimental and clinical VL. Leishmania derived LPG alters the migration of monocytes by decreasing the expression of adhesion molecules and inhibiting the induction and release of MCP-1 [71]. The expression of the genes encoding RANTES, MIP-1α, MIP-1β, CXCL10 and MCP-1 is more strongly upregulated in the air pouch lining of viscerotropic L. donovani-infected animals than in that of dermotropic L. major-infected animals [72], suggesting that leukocyte transendothelial migration could be blocked by L. donovani LPG [73].

4.1.1 Exogenous chemokines host protective immune response in L. donovani infected macrophages:

CC chemokine treatment in the form of MIP-1α and MCP-1 conferred significant protection against L. donovani in an in vitro model of VL [74] by inducing pro-inflammatory cytokines, IL-12 and TNF-α, secretion and inhibiting the anti-inflammatory cytokines, IL-10 and TGF-β, in infected macrophages and also restored the impaired antigen presentation capability of infected macrophages [74, 75] which is critical for generating parasite specific T cell responses [76]. CC

608 ©FORMATEX 2011

Science against microbial pathogens: communicating current research and technological advances A. Méndez-Vilas (Ed.)______________________________________________________________________________

cheomkines strongly induced iNOS2 culminating in a potent generation of NO in L. donovani-infected macrophages and was at par with the concomitant up-regulation of TNF-α release [74]. In addition to NO, MIP-1α and MCP-1 primed parasitized macrophages showed enhanced the chemotactic migration along with significant ROS generation at early time point in L. donovani infected macrophages [77] which were similar to the observation made with antileishmanial drug, sodium stibogluconate [78]. These findings point towards a CC chemokine mediated induction of ROS and NO during early and late phase of infection respectively. Silencing of CCR5, a receptor for MIP-1α, leads to low parasite entry in the macrophages along with enhanced production of NO and proinflammatory cytokines in L. donovani infected macrophages [79]. Comparative study with CXC chemokines, CXCL10 and CXCL8, showed that CXCL10 but not CXCL8, could restrict the intracellular parasitic growth by restoring the impaired proinflammatory cytokines (IL-12 and TNF-α) and chemokines (MIP-1α and MCP-1) along with the NO release in L. donovani infected macrophages [80] which agrees with an earlier finding that Leishmania promastigotes inhibits CXCL10 but induces CXCL8 production [81]. But there was an absence of ROS generation in CXC chemokine primed parasitized macrophages [80] which showcase the difference in the functioning between the CC and CXC chemokines.

4.1.1.1 Exogenous chemokines mediated protection against experimental VL:

In vivo studies with MIP-1α and MCP-1 showed that CC chemokines treatment led to significant decrease in liver and spleen parasite load in infected BALC/c mice [77]. CC chemokine treatment resulted in reduced IL-10 mRNA expression along with enhanced IL-12p40, IFN-γ, TNF-α and iNOS mRNA expression in both liver mononuclear cells as well as in splenocytes, reflecting a switch of CD4+ T cell differentiation from Th2 to Th1 as evident by the increase in IFN-γ secreting CD4+ T cells [75] as host protection is completely dependent on the activation of CD4+ T cells producing Th1-cytokines [82]. Similar to CC chemokines, CXCL10 treatment could also significantly restrict the parasite growth in liver and spleen of L. donovani infected BALB/c mice which were accompanied by a strong induction of Th1 cytokines like IFN-γ and IL-12 along with subsequent abrogation of Th2 cytokines like IL-10 and TGF-β [80, 83]. Additionally, there was a significant increase in IFN-γ secreting CD4+ T cells along with reduction in TGF-β secreting CD4+ CD25+ T regulatory cells in the spleen of CXCL10 treated L. donovani infected mice which is critical for the containment of disease progression [83]. In vivo CXCL10 administration also activates the Natural killer (NK) cells which contribute to promote the protective immune response against Leishmania [84]. The involvement of the CXCR3, receptor for CXCL10, in the disease process was established using CXCR3 (-/-) C57BL/6 mice which show a delayed onset of hepatic inflammation and granuloma formation after L. donovani infection. However, they could mount an efficient Th1 response, recruited T cells to the liver, and controlled parasite growth as efficiently as do CXCR3(+/+) C57BL/6 mice [85].

4.1.1.1.1 Exogenous chemokines rescue the parasite impaired signal transduction during VL:

Treatment with exogenous CC chemokines, MIP-1α and MCP-1, restored the impaired PKC activity during the early hours of L. donovani infection. These chemokines restored Ca2+-dependent PKC activity and inhibited Ca2+-independent atypical PKC activity in L. donovani-infected macrophages under both in vivo and in vitro conditions [77]. This restoration of classical PKC activity by CC chemokines led to ROS generation via membrane translocation of cytosolic factors p47phox and p67phox [77] which are the essential components required for the assembly of an active NADPH oxidase complex [86]. These changes in classical and atypical PKC isoforms was also observed in splenocytes of CC chemokine treated infected mice [77]. On the other hand, exogenous CXC chemokine, CXCL10 antagonistically regulate the MAPK signaling during the disease process. At an early time point, p38MAPK phosphorylation was much higher in CXCL10–treated L. donovani infected macrophages, compared with infected macrophages; in contrast, ERK1/2 phosphorylation in CXCL10–treated L. donovani infected macrophages was much lower than that in infected macrophages [80]. This effective regulation of MAPK signaling by CXCL10 was responsible for enhanced iNOS2 induction and NO production [80]. These findings highlight the importance of PKC and MAPK signaling in the antileishmanial functioning of exogenous CC and CXC chemokines.

5. Conclusion:

These studies on leishmaniasis emphasize on the therapeutic role of exogenous CC and CXC chemokines and their receptors which could regulate the host immunity thereby conferring protection. Further research needs to be undertaken to investigate the role of other novel chemokines in restricting Leishmania induced pathogenesis either alone or in combination with standard antileishmanial drugs.

609©FORMATEX 2011

Science against microbial pathogens: communicating current research and technological advances A. Méndez-Vilas (Ed.)_______________________________________________________________________________

Reference:

[1] Pearson, RD. et al. (2000) Leishmania species: visceral (kala-azar), cutaneous, and mucosal leishmaniasis. In Principles and Practice of Infections Diseases (Mandell, G.L. et al., eds), Churchill Livingston; 2001.

[2] Desjeux P. Human leishmaniases: epidemiology and public health aspects. World Health Stat Q. 1992; 45:267-275. [3] Sacks DL, Perkins PV. Development of infective stage Leishmania promastigotes within phlebotomine sand flies. Am J

Trop Med Hyg. 1985; 34:456-459. [4] Alexander J, Satoskar AR, Russell DG. Leishmania species: models of intracellular parasitism. J Cell Sci. 1999; 112:2993-

3002. [5] Awasthi A, Mathur RK, Saha B. Immune response to Leishmania infection. Indian J Med Res.2004; 119:238-258. [6] Russell DG, Wilhelm H. The involvement of the major surface glycoprotein (gp63) of Leishmania promastigotes in

attachment to macrophages. J Immunol. 1986; 136:2613-2620. [7] 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. J Exp Med. 1995; 181:685-697.

[8] Chan J, Fujiwara T, Brennan P et al. Microbial glycolipids: possible virulence factors that scavenge oxygen radicals. Proc Natl Acad Sci U S A.1989; 86:2453-2457.

[9] Channon JY, Roberts MB, Blackwell JM. A study of the differential respiratory burst activity elicited by promastigotes and amastigotes of Leishmania donovani in murine resident peritoneal macrophages. Immunology.1984; 53:345-355.

[10] Murray HW. Cell-mediated immune response in experimental visceral leishmaniasis. II. Oxygen-dependent killing of intracellular Leishmania donovani amastigotes. J Immunol. 1982; 129: 351-357.

[11] Miller MA, McGowan SE, Gantt KR et al. Inducible resistance to oxidant stress in the protozoan Leishmania chagasi. J Biol Chem. 2000; 275:33883-33889 (2000).

[12] 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. Proc Natl Acad Sci U S A.1992;89:7481-7485.

[13] Buchmüller-Rouiller Y, Mauël J. Impairment of the oxidative metabolism of mouse peritoneal macrophages by intracellular Leishmania spp. Infect Immun.1987; 55:587-593.

[14] Descoteaux A, Turco SJ. Glycoconjugates in Leishmania infectivity. Biochim Biophys Acta.1999; 1455:341-352. [15] Sørensen AL, Hey AS, Kharazmi A. Leishmania major surface protease Gp63 interferes with the function of human

monocytes and neutrophils in vitro. APMIS.1994; 102:265-271. [16] Bhattacharyya S, Ghosh S, Dasgupta B, Mazumder D, Roy S, Majumdar S. Chemokine-induced leishmanicidal activity in

murine macrophages via the generation of nitric oxide. J Infect Dis.2002; 185:1704-1708. [17] Cunha FQ, Assreuy J, Moncada S, Liew FY. Phagocytosis and induction of nitric oxide synthase in murine macrophages.

Immunology. 1993; 79:408-411. [18] Diefenbach A, Schindler H, Donhauser N, Lorenz E, Laskay T, MacMicking J, Röllinghoff M, Gresser I, Bogdan C. 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.

[19] Woods ML, Mayer J, Evans TG, Hibbs JB. Antiparasitic effects of nitric oxide in an in vitro murine model of Chlamydia trachomatis infection and an in vivo murine model of Leishmania major infection. Immunol Ser.1994; 60: 179-195.

[20] Proudfoot L, O'Donnell CA, Liew FY. Glycoinositolphospholipids of Leishmania major inhibit nitric oxide synthesis and reduce leishmanicidal activity in murine macrophages. Eur J Immunol. 1995; 25:745-750.

[21] Proudfoot L, Nikolaev AV, Feng GJ, Wei WQ, Ferguson MA, Brimacombe JS, Liew FY. Regulation of the expression of nitric oxide synthase and leishmanicidal activity by glycoconjugates of Leishmania lipophosphoglycan in murine macrophages. Proc Natl Acad Sci USA. 1996; 93:10984-9.

[22] Brodskyn C, de Oliveira CI, Barral A, Barral-Netto M. Vaccines in leishmaniasis: advances in the last five years. Expert Rev Vaccines. 2003; 2: 705-717.

[23] Sacks D, Noben-Trauth N. The immunology of susceptibility and resistance to Leishmania major in mice. Nat Rev Immunol. 2002; 2: 845-858.

[24] Ohmori Y, Wyner L, Narumi S, Armstrong D, Stoler M, Hamilton TA. Tumor necrosis factor-alpha induces cell type and tissue-specific expression of chemoattractant cytokines in vivo. Am J Pathol. 1993; 142:861-870.

[25] Arnoldi J, Moll H. Langerhans cell migration in murine cutaneous leishmaniasis: regulation by tumor necrosis factor alpha, interleukin-1 beta, and macrophage inflammatory protein-1 alpha. Dev Immunol. 1998; 6:3-11.

[26] Zaph C, Scott P. Interleukin-12 regulates chemokine gene expression during the early immune response to Leishmania major. Infect Immun. 2003; 71:1587-1589.

[27] Farber JM. Mig and IP-10: CXC chemokines that target lymphocytes. J Leukoc Biol, 1997; 61:246-257. [28] Bonecchi R, Bianchi G, Bordignon PP, D'Ambrosio D, Lang R, Borsatti A, Sozzani S, Allavena P, Gray PA, Mantovani A,

Sinigaglia F. Differential expression of chemokine receptors and chemotactic responsiveness of type 1 T helper cells (Th1s) and Th2s. J Exp Med.1998; 187:129-134.

[29] Orlofsky A, Wu Y, Prystowsky MB. Divergent regulation of the murine CC chemokine C10 by Th(1) and Th(2) cytokines. Cytokine.2000; 12:220-228.

[30] Greenberg S. Signal transduction of phagocytosis. Trends Cell Biol.1995; 5:93-99. [31] Bhattacharyya S, Ghosh S, Sen P, Roy S, Majumdar S. Selective impairment of protein kinase C isotypes in murine

macrophage by Leishmania donovani. Mol Cell Biochem. 2001; 216:47-57. [32] Mellor H, Parker PJ. The extended protein kinase C superfamily. Biochem J. 1998; 332: 281-292. [33] Mathur RK, Awasthi A, Wadhone P, Ramanamurthy B, Saha B. Reciprocal CD40 signals through p38MAPK and ERK-1/2

induce counteracting immune responses. Nat Med. 2004; 10:540-544.

610 ©FORMATEX 2011

Science against microbial pathogens: communicating current research and technological advances A. Méndez-Vilas (Ed.)______________________________________________________________________________

[34] Su B, Karin M. Mitogen-activated protein kinase cascades and regulation of gene expression. Curr Opin Immunol.1996; 8:402-411.

[35] Seger R, Krebs EG. The MAPK signaling cascade. FASEB J. 1995; 9:726-735. [36] Salmon RA, Guo X, Teh HS, Schrader JW. The p38 mitogen-activated protein kinases can have opposing roles in the

antigen-dependent or endotoxin-stimulated production of IL-12 and IFN-gamma. Eur J Immunol.2001; 31:3218-3227. [37] Ajizian SJ, English BK, Meals EA. Specific inhibitors of p38 and extracellular signal-regulated kinase mitogen-activated

protein kinase pathways block inducible nitric oxide synthase and tumor necrosis factor accumulation in murine macrophages stimulated with lipopolysaccharide and interferon-gamma. J Infect Dis. 1999; 179:939-944.

[38] Evans TG, Reed SS, Hibbs JB. Nitric oxide production in murine leishmaniasis: correlation of progressive infection with increasing systemic synthesis of nitric oxide. Am J Trop Med Hyg. 1996; 54:486-489.

[39] Awasthi A, Mathur R, Khan A Joshi BN, Jain N, Sawant S, Boppana R, Mitra D, Saha B. CD40 signaling is impaired in L. major-infected macrophages and is rescued by a p38MAPK activator establishing a host-protective memory T cell response. J Exp Med. 2003; 197:1037-1043.

[40] Nishizuka Y. The molecular heterogeneity of protein kinase C and its implications for cellular regulation. Nature. 1998; 334:661-665.

[41] Giorgione JR, Turco SJ, Epand RM. Transbilayer inhibition of protein kinase C by the lipophosphoglycan from Leishmania donovani. Proc Natl Acad Sci USA.1996; 93:11634-11639.

[42] Mukhopadhyay S, Sen P, Bhattacharyya S, Majumdar S, Roy S. Immunoprophylaxis and immunotherapy against experimental visceral leishmaniasis. Vaccine.1999; 17:291-300.

[43] Allen LA, Aderem A. Molecular definition of distinct cytoskeletal structures involved in complement- and Fc receptor-mediated phagocytosis in macrophages. J Exp Med. 1996; 184:627-637.

[44] Allen LH, Aderem A. A role for MARCKS, the alpha isozyme of protein kinase C and myosin I in zymosan phagocytosis by macrophages. J Exp Med.1995; 182:829-840.

[45] 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. Infect Immun. 2001; 69:1499-1507.

[46] Melby, PC, Valencia-Pacheco G, Andrade-Narváez F. Induction of macrophage killing of intracellular Leishmania donovani by human T cell clones. Arch Med Res. 1996; 27:473-479.

[47] Moser B, Wolf M, Walz A, Loetscher P. Chemokines: multiple levels of leukocyte migration control. Trends Immunol. 2004;25:75-84.

[48] Ward SG, Bacon K, Westwick J. Chemokines and T lymphocytes: more than an attraction. Immunity. 1998; 9:1-11. [49] Luster AD. Chemokines--chemotactic cytokines that mediate inflammation. N Engl J Med. 1998; 338: 436-445. [50] Rot A, von Andrian UH. Chemokines in innate and adaptive host defense: basic chemokinese grammar for immune cells.

Annu Rev Immunol. 2004; 22:891-928. [51] Viola A, Luster AD. Chemokines and their receptors: drug targets in immunity and inflammation. Annu Rev Pharmacol

Toxicol. 2008; 48:171-197. [52] Baggiolini M, Dewald B, Moser B. Human chemokines: an update. Annu Rev Immunol. 1997; 15: 675-705. [53] Premack BA, Schall TJ. Chemokine receptors: gateways to inflammation and infection. Nat Med. 1996; 2:1174-1178. [54] Del Rio L, Bennouna S, Salinas J, Denkers EY. CXCR2 deficiency confers impaired neutrophil recruitment and increased

susceptibility during Toxoplasma gondii infection. J Immunol. 2001; 167:6503-6509. [55] Jones E, Price DA, Dahm-Vicker M, Cerundolo V, Klenerman P, Gallimore A. The influence of macrophage inflammatory

protein-1alpha on protective immunity mediated by antiviral cytotoxic T cells. Immunology.2003; 109:68-75. [56] Rodriguez-Sosa M, Rosas LE, Terrazas LI, Lu B, Gerard C, Satoskar AR. CC chemokine receptor 1 enhances susceptibility

to Leishmania major during early phase of infection. Immunol Cell Biol. 2003; 81:114-120. [57] Caswell JL, Middleton DM, Sorden SD, Gordon JR. Expression of the neutrophil chemoattractant interleukin-8 in the

lesions of bovine pneumonic pasteurellosis. Vet Pathol. 1998; 35:124-131. [58] Entrican G, Wilkie R, McWaters P, Scheerlinck J, Wood PR, Brown J. Cytokine release by ovine macrophages following

infection with Chlamydia psittaci. Clin Exp Immunol.1999; 117: 309-315. [59] Zhang Y, Broser M, Cohen H, Bodkin M, Law K, Reibman J, Rom WN. Enhanced interleukin-8 release and gene

expression in macrophages after exposure to Mycobacterium tuberculosis and its components. J Clin Invest. 1995; 95:586-592.

[60] Majumder N, Bhattacharjee S, Bhattacharyya Majumdar S, Dey R, Guha P, Pal NK, Majumdar S. Restoration of impaired free radical generation and proinflammatory cytokines by MCP-1 in mycobacterial pathogenesis. Scand J Immunol. 2008; 67:329-339.

[61] Balamayooran G, Batra S, Balamayooran T, Cai S, Jeyaseelan S. Monocyte Chemoattractant Protein 1 Regulates Pulmonary Host Defense via Neutrophil Recruitment during Escherichia coli Infection. Infect Immun. 2011; 79: 2567-2577.

[62] Furie MB, Randolph GJ. Chemokines and tissue injury. Am J Pathol. 1995; 146:1287-1301. [63] Cook DN, Beck MA, Coffman TM et al. Requirement of MIP-1 alpha for an inflammatory response to viral infection.

Science. 1995; 269:1583-1585. [64] Cocchi F, DeVico AL, Garzino-Demo A, Arya SK, Gallo RC, Lusso P. Identification of RANTES, MIP-1 alpha, and MIP-

1 beta as the major HIV-suppressive factors produced by CD8+ T cells. Science. 1995; 270: 1811-1815 (1995). [65] Villalta F, Zhang Y, Bibb KE, Kappes JC, Lima MF. The cysteine-cysteine family of chemokines RANTES, MIP-1alpha,

and MIP-1beta induce trypanocidal activity in human macrophages via nitric oxide. Infect Immun. 1998; 66:4690-4695. [66] Krathwohl MD, Anderson JL. Chemokine CXCL10 (IP-10) is sufficient to trigger an immune response to injected antigens

in a mouse model. Vaccine. 2006; 24:2987-2993.

611©FORMATEX 2011

Science against microbial pathogens: communicating current research and technological advances A. Méndez-Vilas (Ed.)_______________________________________________________________________________

[67] Cotterell SE, Engwerda CR, Kaye PM. Leishmania donovani infection initiates T cell-independent chemokine responses, which are subsequently amplified in a T cell-dependent manner. Eur J Immunol. 1999; 29: 203-214.

[68] Ato M, Stäger S, Engwerda CR, Kaye PM. Defective CCR7 expression on dendritic cells contributes to the development of visceral leishmaniasis. Nat Immunol. 2002; 3:1185-1191.

[69] Sato N, Kuziel WA, Melby PC, Reddick RL, Kostecki V, Zhao W, Maeda N, Ahuja SK, Ahuja SS. Defects in the generation of IFN-gamma are overcome to control infection with Leishmania donovani in CC chemokine receptor (CCR) 5-, macrophage inflammatory protein-1 alpha-, or CCR2-deficient mice. J Immunol. 1999; 163: 5519-5525.

[70] Hailu A, van der Poll T, Berhe N, Kager PA. Elevated plasma levels of interferon (IFN)-gamma, IFN-gamma inducing cytokines, and IFN-gamma inducible CXC chemokines in visceral leishmaniasis. Am J Trop Med Hyg. 2004; 71:561-567.

[71] Sticherling M, Küpper M, Koltrowitz F, Bornscheuer E, Kulke R, Klinger M, Wilhelm D, Kameyoshi Y, Christophers E, Schröder JM. Detection of the chemokine RANTES in cytokine-stimulated human dermal fibroblasts. J Invest Dermatol. 1995; 105: 585-591.

[72] Matte C, Olivier M. Leishmania-induced cellular recruitment during the early inflammatory response: modulation of proinflammatory mediators. J Infect Dis. 2002; 185: 673-681.

[73] Lo SK, Bovis L, Matura R, Zhu B, He S, Lum H, Turco SJ, Ho JL. Leishmania lipophosphoglycan reduces monocyte transendothelial migration: modulation of cell adhesion molecules, intercellular junctional proteins, and chemoattractants. J Immunol. 1998; 160: 1857-1865.

[74] Bhattacharyya S, Ghosh S, Dasgupta B, Mazumder D, Roy S, Majumdar S. Chemokine-induced leishmanicidal activity in murine macrophages via the generation of nitric oxide. J Infect Dis, 185(12), 1704-1708 (2002).

[75] Dey R, Majumder N, Bhattacharyya Majumdar S, Bhattacharjee S, Banerjee S, Roy S, Majumdar S. Induction of host protective Th1 immune response by chemokines in Leishmania donovani-infected BALB/c mice. Scand J Immunol. 2007;66:671-83.

[76] Pinelli E, Rutten VP, Bruysters M, Moore PF, Ruitenberg EJ. Compensation for decreased expression of B7 molecules on Leishmania infantum-infected canine macrophages results in restoration of parasite-specific T-cell proliferation and gamma interferon production. Infect Immun. 1999; 67:237-243.

[77] Dey R, Sarkar A, Majumder N Bhattacharyya Majumdar S, Roychoudhury K, Bhattacharyya S, Roy S, Majumdar S. Regulation of impaired protein kinase C signaling by chemokines in murine macrophages during visceral leishmaniasis. Infect Immun. 2005; 73:8334-8344.

[78] Rais S, Perianin A, Lenoir M, Sadak A, Rivollet D, Paul M, Deniau M. Sodium stibogluconate (Pentostam) potentiates oxidant production in murine visceral leishmaniasis and in human blood. Antimicrob Agents Chemother. 2000; 44:2406-2410.

[79] Bhattacharyya S, Dey R, Majumder N, Bhattacharjee S, Majumdar S. A novel approach to regulate experimental visceral leishmaniasis in murine macrophages using CCR5 siRNA. Scand J Immunol. 2008; 67:345-53.

[80] Gupta G, Bhattacharjee S, Bhattacharyya S, Bhattacharya P, Adhikari A, Mukherjee A, Bhattacharyya Majumdar S, Majumdar S. CXC chemokine-mediated protection against visceral leishmaniasis: involvement of the proinflammatory response. J Infect Dis.2009; 200:1300-10.

[81] 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. Infect Immun. 2002; 70:4177-84.

[82] Scott P, Artis D, Uzonna J, Zaph C. The development of effector and memory T cells in cutaneous leishmaniasis: the implications for vaccine development. Immunol Rev.2004; 201:318-38.

[83] Gupta G, Majumdar S, Adhikari A, Bhattacharya P, Mukherjee AK, Majumdar SB, Majumdar S. Treatment with IP-10 induces host-protective immune response by regulating the T regulatory cell functioning in Leishmania donovani-infected mice. Med Microbiol Immunol.2011; 200: 241-53.

[84] Vester B, Müller K, Solbach W, Laskay T. Early gene expression of NK cell-activating chemokines in mice resistant to Leishmania major. Infect Immun. 1999; 67:3155-3159.

[85] Barbi J, Oghumu S, Rosas LE Carlson T, Lu B, Gerard C, Lezama-Davila CM, Satoskar AR. Lack of CXCR3 delays the development of hepatic inflammation but does not impair resistance to Leishmania donovani. J Infect Dis. 2007; 195: 1713-1717.

[86] DeLeo FR, Quinn MT. Assembly of the phagocyte NADPH oxidase: molecular interaction of oxidase proteins. J Leukoc Biol.1996; 60:677-691.

612 ©FORMATEX 2011

Science against microbial pathogens: communicating current research and technological advances A. Méndez-Vilas (Ed.)______________________________________________________________________________