Next Article in Journal
Schistosomiasis at the Crossroad to Elimination: Review of Eclipsed Research with Emphasis on the Post-Transmission Agenda
Next Article in Special Issue
Cubital Tunnel Syndrome Temporally after COVID-19 Vaccination
Previous Article in Journal
Transmission of SARS-CoV-2 in the Population Living in High- and Low-Density Gradient Areas in Dhaka, Bangladesh
Previous Article in Special Issue
Diagnostic Value of Neutrophil-to-Lymphocyte Ratio, Lymphocyte-to-Monocyte Ratio, and Platelet-to-Lymphocyte Ratio in the Diagnosis of Erythema Nodosum Leprosum: A Retrospective Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Responses in Leishmaniasis: An Overview

by
Ana Caroline Costa-da-Silva
1,
Danielle de Oliveira Nascimento
2,
Jesuino R. M. Ferreira
3,
Kamila Guimarães-Pinto
3,
Leonardo Freire-de-Lima
1,
Alexandre Morrot
4,5,
Debora Decote-Ricardo
2,
Alessandra Almeida Filardy
3 and
Celio Geraldo Freire-de-Lima
1,*
1
Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21944-970, Brazil
2
Instituto de Veterinária, Universidade Federal Rural do Rio de Janeiro, Seropédica 23890-000, Brazil
3
Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21944-970, Brazil
4
Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-900, Brazil
5
Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21045-900, Brazil
*
Author to whom correspondence should be addressed.
Trop. Med. Infect. Dis. 2022, 7(4), 54; https://doi.org/10.3390/tropicalmed7040054
Submission received: 14 February 2022 / Revised: 24 March 2022 / Accepted: 29 March 2022 / Published: 31 March 2022
(This article belongs to the Special Issue Feature Papers in Tropical Medicine and Infectious Disease)

Abstract

:
Leishmaniasis is a parasitic, widespread, and neglected disease that affects more than 90 countries in the world. More than 20 Leishmania species cause different forms of leishmaniasis that range in severity from cutaneous lesions to systemic infection. The diversity of leishmaniasis forms is due to the species of parasite, vector, environmental and social factors, genetic background, nutritional status, as well as immunocompetence of the host. Here, we discuss the role of the immune system, its molecules, and responses in the establishment, development, and outcome of Leishmaniasis, focusing on innate immune cells and Leishmania major interactions.

1. Leishmaniasis

Leishmaniasis is a neglected endemic tropical disease distributed in more than 90 countries throughout the New World (Latin America) and Old World (Africa, Asia, and Southern Europe), primarily found in Southeast Asia, East Africa, and Brazil. It has an estimated prevalence of 12 million cases worldwide, which is continuously growing, with 1.5–2 million new cases each year [1,2]. Leishmaniasis is an infectious disease caused by parasites of the genus Leishmania, of the Trypanosomatidae family. This disease manifests in four main forms: cutaneous leishmaniasis (CL), diffuse cutaneous leishmaniasis (DCL), mucocutaneous leishmaniasis (MCL), and visceral leishmaniasis (VL) or kala-azar. CL, the most common form, causes non-lethal skin lesions and is primarily caused by L. major, L. tropica, and L. aethiopica in the Old World, or by L. mexicana, L. amazonensis, L. braziliensis, L. panamensis, and L. guyanensis in the New World [3]. DCL, the anergic form of CL, is a rare condition in which lesions are full of parasites, and is characterized by multiple nodules, papules, or tubercles with diffuse skin infiltration and no ulceration. The main causative species of DCL are L. mexicana and L. amazonensis in the New World; and L. aethiopica in the Old World. VL, the most severe clinical form of leishmaniasis, is responsible for thousands of deaths each year and is caused by L. donovani and L. infantum in the Old World or L. chagasi; and by L. infantum in the New World. This form is characterized by prolonged fever, splenomegaly, hypergammaglobulinemia, and pancytopenia. Finally, MCL is mainly caused by L. braziliensis and eventually by L. panamensis or L. guyanensis. About 90% of MCL cases occur in Bolivia, Brazil, and Peru. This form of leishmaniasis is characterized by the dissemination of the infection from primary cutaneous lesions to the mucosal system via direct extension, bloodstream, or lymphatics [4,5,6].
The transmission of the protozoan to humans or other mammals occurs through the bite of the female sandfly belonging to the order Diptera, family Psychodidae, and subfamily Phlebotominae of the genera Phlebotomus (Old World) and Lutzomyia (New World). Leishmania has a relatively simple heteroxenic life cycle: an extracellular promastigote stage, which can be either procyclic, which multiplies and develops in the digestive tract of sandflies, or metacyclic infective, which migrates to the proboscis of the sandfly to be inoculated during the blood meal; as well as an intracellular stage in the form of a spherical, immobile amastigote, which is morphologically and biochemically distinct from promastigotes, and resides and multiplies in the phagolysosomes of phagocytes in their vertebrate hosts [7,8].
Recent advances in imaging technologies, as well as studies combining genetic and immunological manipulations, have allowed a better understanding of the interactions between Leishmania and its mammalian host, especially the role of different cell types involved in the initiation and development of the immune (Figure 1) [7,9]. The divergent clinical manifestations observed in leishmaniasis are dependent not only on the genetic background, nutritional status, and immunocompetence of the host, but also largely on the species of parasite that initiates the infection, the vector, and environmental and social factors [10].

2. Th1 versus Th2 Response to Leishmania Infection

The balance between the type 1 and type 2 responses, along with regulatory mechanisms, is a determinant of the outcome of leishmaniasis. The L. major mouse infection model has been widely used to decipher some of the immune response mechanisms involved in susceptibility and resistance to a parasitic infection, even though it does not apply consistently to all forms of leishmaniasis.
The cellular immunity generated by a Th1/Tc1 response is considered an important mediator of resistance to Leishmania. Subcutaneous infection with L. major promastigotes promotes a Th1 skewed protective response in resistant murine strains, such as C57BL/6, C3H, and CBA, leading to the development of small lesions that heal spontaneously, control of parasite replication, and immunity against reinfection [11,12,13]. On the other hand, an improperly modulated and exacerbated type 1 response can result in severe tissue damage and clinical presentation of leishmaniasis. CL caused by L. tropica [14] and MCL caused by L. braziliensis and L. amazonensis [15] are characterized by increased amounts of proinflammatory cytokines, including IFNγ and TNFα.
With regard to susceptible murine strains, such as BALB/c, the course of L. major infection is characterized by the development of a Th2 response with persistent inflammatory lesions, uncontrolled replication of the parasite, and its systemic dissemination to lymph nodes and the spleen [11,12,13]. Interestingly, a previous study demonstrated that Balb/c mice lacking IL-4Rα signaling in dendritic cells (DCs) are highly susceptible to L. major infection, pointing to the role of early IL-4R signaling on DCs for protection against CL [16].
In addition to Th1 and Th2 responses, other mechanisms are described as important for disease control or progression, depending on the species of Leishmania and the animal model employed. Increased levels of IL-17 as well as IL-10 have been described in patients with CL and MCL [17,18,19], implying the role of Th17 cells and regulatory T cells (Tregs) in the pathogenesis of leishmaniasis. Nonetheless, the role of these cells has been thoroughly discussed by others [20,21,22], and is not the scope of this review.

3. Innate Immune Response to Leishmania Infection

The immune response is initiated at the site of pathogen entry. Upon inoculation of Leishmania in the dermis, promastigotes interact with serum components, activating the complement system, both classical and alternative pathways. The opsonization of metacyclic promastigote forms by complement is rapid and efficient, resulting in approximately 90% lysis of the inoculated parasites. However, the parasites developed mechanisms that allowed them to resist and bypass this lysis process [23,24,25,26]. First, promastigote metacyclogenesis gives rise to more complement-resistant forms. Furthermore, Leishmania expresses protein kinases that phosphorylate C3, C5, and C9 components, inhibiting complement activation [27,28]. Finally, two molecules on the surface of the parasite, lipophosphoglycan (LPG) and glycoprotein of 63 kDa (GP63), mediate their binding to inactivated C3b (iC3b), preventing complement-mediated lysis, and facilitating the internalization of Leishmania via complement receptors (CRs) [29].
Sandfly saliva also contributes to the infection outcome. It contains exosomes [30], gut microbes [31], and molecules that have diverse activities including vasodilation, coagulation inhibition, and immunomodulatory effects [32]. Furthermore, it has already been described that the promastigote forms of Leishmania induce the secretion of MCP-1, CXCL1, and CXCL2 which act by attracting both monocytes and neutrophils [33,34]. Indeed, sustained recruitment of phagocytic cells to the bite site plays an essential role in infection establishment.
Resident cells, such as macrophages, keratinocytes, mast cells, and Langerhans cells, called sentinel and present at the inoculation site, express a variety of pattern recognition receptors (PRRs), including Toll-like receptors (TLRs), which recognize pathogen-associated molecular patterns (PAMPs) and trigger the phagocytosis of microorganisms and opsonized particles. These cells also express several cytokine receptors and, together with other tissue cells, produce several chemokines, initiating innate and acquired immune response activation cascades [33,35].
Activated cells undergo several morphological and functional changes that lead to parasite phagocytosis and oxidative stress. After their initial encounter with neutrophils, Leishmania parasites are phagocytosed by both monocytes and macrophages, and DCs [36]. Phagocytosis involves three distinct events: binding to specific receptors, engulfment with the formation of the parasitophorous vacuole, and parasite death or degradation by the formation of reactive oxygen species/reactive nitrogen species (ROS/RNS) [37,38]. These specific events are discussed further in a later section.

3.1. Neutrophils–Leishmania Interaction

Neutrophils are the first cells to arrive at the site of Leishmania infection and their role can be either beneficial or detrimental, depending on the Leishmania species and host factors [39]. Interestingly, in the murine model of CL, recruitment of neutrophils depends on whether the mouse strain is susceptible (BALB/c) or resistant (C57BLl/6) to the infection. Initially, both strains show a similar inflammatory response. However, neutrophils become prominent and persist in susceptible hosts, whereas this recruitment is not sustained in resistant animals, returning to basal levels within three days [11,40,41].
Infiltrating neutrophils target and eliminate leishmanial parasites through different mechanisms, including the phagocytosis and production of an array of intracellular and extracellular microbicidal factors such as ROS and neutrophil extracellular traps (NETs) [24,42]. However, Leishmania can survive transiently within neutrophils through several protective mechanisms, including the inhibition of phagolysosome biogenesis [43], the prevention of oxidative stress [44], and the delay in neutrophil apoptosis [45]. Besides, infected neutrophils also induce the production of a variety of chemokines and cytokines, such as interleukin (IL)-8 and MIP1β, which attract additional neutrophils and other phagocytic cells, favoring Leishmania survival and pathology [46,47,48].
In vitro studies suggest that Leishmania uses neutrophils as intermediary cells for the infection of its definitive host cell, in a model called “Trojan Horse” [47,49]. In this model, macrophages are able to phagocytose parasitized neutrophils, and the parasites survive and multiply within the macrophage vacuoles, even managing to multiply within macrophages. Phagocytosis of dead neutrophils can be both pro- and anti-inflammatory. In susceptible mice, such as BALB/c, phagocytic removal of apoptotic neutrophils (efferocytosis) exacerbates the growth of the parasite inside macrophages through the production of TGFβ and PGE2 [50,51]. Furthermore, the depletion of neutrophils inhibits the production of IL-4 and reduces the parasite’s ability to engage in productive cycles of infection and parasite burden, providing a better response to infection [40,49]. In contrast, the phagocytic removal of apoptotic neutrophils by macrophages from resistant C57BL/6 mice induces the death of the parasite mediated by the enhanced production of neutrophilic elastase and TNFα [52]. The transient depletion of these neutrophils at the time of infection, in turn, increases the growth of the parasite and the size of the lesion in resistant mice, even if, in this case, these mice eventually heal [40,50]. The definitive role of neutrophils in Leishmania infection outcome has yet to be defined with more comprehensive models employing different parasite species and genetic backgrounds.

3.2. Macrophage–Leishmania Interaction

The interaction between macrophages and Leishmania plays a fundamental role in the pathogenesis of the infection. In the experimental leishmaniasis models, macrophages are crucial cells not only for the survival, replication, and differentiation of the parasites, but also for their elimination [53]. Macrophages correspond to the main reservoir of Leishmania in vivo. Although it has already been demonstrated that these cells are not capable of producing IL-12 after infection by L. major [54], macrophages are capable of producing pro-inflammatory cytokines at the site of infection and killing and eliminating parasites through the production of nitric oxide (NO) [55]. They also induce the recruitment of pro-inflammatory cells, in addition to presenting Leishmania antigens to the T cells, along with DCs for primed T cells.

3.3. Dendritic Cells–Leishmania Interaction

DCs play an essential role in the initiation and regulation of effective immune responses against Leishmania. These cells can uptake and process antigens; mature and upregulate MHCII and co-stimulatory molecules; migrate to lymph nodes; and activate T cells to differentiate into effector Th1 cells through the production of IL-12 and IL-27 [56,57,58]. Distinct DCs subsets have been identified in the skin and several studies have shown their different functions in T cell activation and disease outcome.
Langerhans cells (LCs) were initially considered essential for the control of cutaneous leishmaniasis [59]. However, later studies demonstrated that these cells were not required for the generation of protective immunity against Leishmania parasites [60]. In fact, it was demonstrated that these cells can have a regulatory function, once the depletion of LCs dampens the production of IL-10 and the numbers of Tregs and favors the production of IFNγ and the decrease in parasite load [61]. Instead of LCs, the participation of dermal DCs (CD11c+ CD8α Langherin) and monocyte-derived DCs proved to be essential for the transport of Leishmania antigens from the infection site to the draining lymph node and in the induction of a specific T cell response [62,63]. Interestingly, IL-4, a type 2 cytokine, is required for the optimal induction of Th1 responses by DCs as it inhibits IL-10 production [64,65].

3.4. Leishmania Interaction with Other Innate Immune Cells

Dendritic cells are not the only cell type needed and involved in developing an effective T cell response. Some studies demonstrate that natural killer (NK) cells have a protective role in leishmaniasis, being the primary source of IFNγ for the development of a Th1-type response [66,67]. Indeed, reduced numbers of NK cells and activation markers, such as IFNγ and TNFα, were observed in patients with diffuse cutaneous leishmaniasis compared to localized CL [66]. Recently, a contribution of NK cells to the immunopathology of CL was also demonstrated mainly through the release of granzyme B. It was suggested that NK cells can contribute to cytotoxicity activity in CL patients [68].
Mast cells are also present at the site of parasite inoculation and seem to be important to the outcome of leishmaniasis. These cells produce a variety of mediators and cytokines, participating in several stages of the innate and adaptive immune response. However, the precise role of these cells in Leishmania infection is not well understood [69]. Infection of mast cell-deficient C57BLl/6 mice with L. major demonstrated a protective role for these cells, since their absence led to the development of larger skin lesions containing greater amounts of parasites and also an increase in the spread of these parasites to the spleen [70,71]. On the other hand, other studies suggest that mast cells play a deleterious role in Leishmania infection, through the production of IL-4 and IL-13 and the establishment of Th2 response [72,73]. Furthermore, mast cells’ degranulation before L. major infection showed their ability to regulate not only inflammation through histamine release, but also to promote Th2 response [74].
Keratinocytes have already been described as having an important role in the initiation of infection, acting not only as a physical barrier, but also secreting molecules that can shape the immune responses to the pathogen. For example, it has already been reported that keratinocytes are capable of secreting immunomodulatory mediators such as IL-12, IL-1β, osteopontin, IL-4, and IL-6 [75].

4. Leishmania and Host Immune Receptors’ Interaction and Intracellular Processing of Leishmania

The first crucial event for Leishmania infection involves the initial contact and stable interaction between the promastigote forms of the parasite and host cells. Recognition and phagocytose of promastigotes can be mediated by several molecules including CR1, CR3 (Mac-1), fibronectin receptor (FnR) and C-type lectin receptors (CLRs) expressed on the surface of macrophages [74]. However, it has been demonstrated that the individual binding of these receptors does not lead to macrophage activation, suggesting that multiple receptors may be important for the initiation of the appropriate immune response [75].
The interaction of Leishmania with macrophages primarily occurs through CR1 and CR3 receptors that recognize iC3b-opsonized parasites [76,77]. Inactivation of C3b molecules is a defense mechanism developed by Leishmania that allows its silent entry into macrophages with no induction of oxidative stress [77], and reduced production of IL-12 [78]. Mechanisms of C3 molecule inactivation have been attributed to the protease gp63 [79] and uptake of factor H, a regulatory molecule of the complement [26].
FnR cooperates with CR3 for binding and phagocytosis of promastigotes. Previous studies demonstrated that this receptor increases Leishmania binding to phagocytes [80,81,82], but, interestingly, FnR also decreases Leishmania intracellular survival [82].
Some conflicting data have been observed regarding the participation of CLRs, such as Dectin-1, mannose, and Mincle receptors, in the internalization of promastigote forms of Leishmania by macrophages and dendritic cells [83].
The precise role of Leishmania binding to MR, whether pro- or anti-inflammatory, is still unclear. L. donovani promastigotes’ interaction with monocyte-derived macrophages was partially inhibited by the addition of specific MR ligands and monoclonal antibodies against the receptor [84]. Stimulation of MR promoted an effective immune response and clearance of L. infantum infection [83]. However, the use of MR-deficient animals showed that the phagocytosis of metacyclic promastigotes of both L. major and L. donovani occurred efficiently and similarly to that in wild-type animals [80].
Dectin-1 also promoted oxidative stress production and clearance of Leishmania in a VL murine model [83]. An increased expression of Dectin-1 was also observed after macrophages’ infection with L. amazonensis in vitro [84] and, more recently, the expansion of Dectin-1 + DCs was also observed in experimental leishmaniasis as well as in patients suffering from CL [81].
Importantly, SIGNR3 expression in macrophages favored L. infantum resilience through decreased IL-1β expression, whereas Mincle was shown to promote the infection of L. major, by dampening dendritic cell priming through the activation of the inhibitory ITAM pathway [82].
Despite the large amount of information available about the phagocytosis process of the promastigote forms, few studies have been conducted on the phagocytic processes of the amastigote forms, which are responsible for the maintenance and dissemination of the infection in the vertebrate host. Promastigote forms encounter macrophages only in the early stages of infection, whereas amastigotes are continuously released from infected cells and are internalized by other uninfected cells, giving rise to a progressive infection. In addition, opsonized amastigotes are phagocytosed via FcγR, a process that results in IL-10 secretion, facilitating the survival and replication of the parasite. [85,86,87,88] In fact, BALB/c JHD (deficient in circulating antibodies) or FcγR-/- animals have smaller skin lesions after infection by L. amazonensis and L. pifanoi [89].
It is also worth mentioning that DCs preferentially internalize the amastigotes of the parasite via FcγR [90,91] and, therefore, the infection of these cells occurs in the late stages of infection, when L. major amastigotes are released into the tissue. This scenario provides plausible explanations for the delay that occurs between parasite inoculation and the development of a cellular immune response that is observed in cutaneous leishmaniasis.
In addition to FcRs, other receptors participate in the phagocytosis of amastigotes, such as CRs, FnR, and heparin-binding protein, and it was also seen that binding to the MR, in this case, did not affect the entry of amastigotes [92].
Although surface receptors initially determine the phagocytosis of Leishmania and its route within host cells, the internalized parasites continue to apply strategies to promote their survival. Besides being crucial for the initiation of phagocytosis and subsequent intracellular survival of the parasite, L. major surface molecules such as LPG, gp63, and proteophosphoglycans can also act as ligands for different TLRs [93].
TLRs play an important role in the recognition of pathogens and activation of immune cells. Importantly, TLR2, TLR4, and TLR9 have been described as important mediators of immune cells and Leishmania interaction [94]. Mice lacking TLR signaling are more susceptible to L. major infection, presenting a decreased ability to induce protective immunity and increased parasite burden [95,96]. In contrast, the absence of TLR signaling in mice infected with L. amazonensis resulted in the improvement of immune responses with a higher production of IL-12 and enhanced resistance to infection [97]. More recently, decreased levels of inflammatory molecules and reduced parasitic load were observed after the neutralization of TLR2 and TLR4 in CL patient monocytes [92]. TLR9-dependent activation of macrophages [98], dendritic cells [99], and NK cells [100] is also important for the resolution of Leishmania infection.
After the internalization of the promastigote forms into phagosomes, lysosomes fuse for the complete formation of the parasitophorous vacuole. Walker et al. demonstrated that LPG impairs vacuole acidification by inhibiting the functional assembly of the NADH oxidase complex and preventing vacuolar proton recruitment ATPase on the parasitophorous membrane. This process not only allows the transformation of the parasite into amastigotes [101], but also inhibits the activation of lysosomal proteases necessary for antigen processing and the initiation of the immune response [43].
Unlike promastigotes, amastigotes are able to survive inside the phagolysosome because, among other things, they have proton pumps in their plasma membrane that capture metabolites and also metabolite transporters whose functions are properly exercised at acidic pH [102]. The zinc-metalloprotease GP63, or leishmaniolysin, the most abundant protein in amastigotes, is an endoproteinase whose proteolytic activity at an acidic pH is relevant for the survival of amastigotes in phagolysosomes, probably through the inactivation of lysosomal macrophage proteins [103,104].
In general, binding to specific receptors culminates in the activation of different pathways and functions in macrophages, and each interaction occurs because of factors expressed by the parasite itself. These virulence factors are regulated during the parasite life cycle and guarantee its ability to select the routes of immune system invasion and evasion [92].

5. Changes in Macrophages after Leishmania Infection

Changes in macrophages caused by Leishmania infection have been the subject of several studies to understand how the promastigote forms, after being internalized, are able to differentiate into amastigotes and multiply within a hostile environment such as the parasitophorous vacuole. Studies have shown that macrophages infected with Leishmania have a deficient response to IFNγ, characterizing them as deactivated macrophages [105]. Filardy et al. showed that the infection with L. major rapidly triggers a cellular stress response in residents, but not inflammatory peritoneal macrophages. This response induces proinflammatory signals, such as the secretion of the cytokines/chemokines TNF-α, IL-6, TIMP-1, IL-1RA, GCSF, TREM, KC, MIP-1α, MIP-1β, MCP-1, and MIP-2, but is also involved in parasite survival and replication in host macrophages [38]. Furthermore, the critical involvement of signaling through MyD88 and TLR in protecting against L. major infection is well established [106].
The first study to characterize which genes were modulated by Leishmania infection in macrophages from BALB/c mice revealed that 37% of the analyzed genes, totaling 588 genes, including the CD40 gene, were downregulated in infected macrophages with L. donovani, when compared to non-infected macrophages not infected with L. donovani. In contrast, only eight of the messenger RNAs (mRNA) studied had their expression increased, some of them linked to the recruitment of more macrophages to the site of infection, including MIP-1α and MIP-1β genes [107]. Dillon et al. also aimed to identify global changes in gene expression using murine macrophages from C57BL/6 mice and L. major at 4, 24, 48, and 72 h post-infection [108]. They demonstrated that genes related to both pro- and anti-inflammatory immune responses and glycolysis were substantially upregulated, and genes related to lipid metabolism, biogenesis, and Fc gamma receptor-mediated phagocytosis were downregulated in the murine macrophages. Human monocytes infected with L. major also disclosed an upregulation of pro-inflammatory cytokine and cytokines receptors including IL1A, IL1RN, IL6, and IL6R [109].
Another study comparing L. major and L. donovani infection in human macrophages revealed that the profile of modulated genes differs between species of the same genus, probably resulting in the different clinical manifestations observed. In this study, negative modulation of a group of genes induced by IFNγ was observed in macrophages infected with L. major. This study, however, showed that similar amounts of mRNA were both up- and down-regulated by both Leishmania species [110]. Shadab et al. described that murine peritoneal macrophages infected with virulent L. donovani strains showed suppression of many important cellular processes, including protein synthesis, in comparison to non-virulent variants. Genes encoding virulence factors and those important for parasite survival were significantly upregulated in the intracellular virulent amastigotes. In contrast, genes involved in the immune stimulations and negative regulation of the cell cycle and transcriptional regulation were also all upregulated in the non-virulent strains [111].
Rodriguez et al. demonstrated in a model of macrophage infection of BALB/c animals with L. chagasi the negative modulation of several pro-inflammatory genes, such as phagocytic receptors linked to classical macrophage activation—FcγRI and FcγRIIb. Furthermore, positive modulation of several genes associated with alternative activation of macrophages or Th2 response was observed, such as MR, CCR3, MIP-2, and TGF-βRII, among others [105].
Transcriptional analysis of macrophages from BALB/c mice infected with L. amazonensis amastigotes was also performed and the authors observed the induction of genes related to alternative macrophage activation, such as the increased expression of the arginase 2 gene, as well as IL-1Ra and the reduced expression of the CD14 gene [112]. Corroborating this work, the results of an experimental system with dual RNA sequencing of enucleated fibroblasts (cytoplasts) and intracellular L. amazonensis, which was performed recently to obtain further insights into parasites’ control over the host cell, suggested that a parasite-mediated control of the host cell transcripts’ half-life was beneficial to the parasite’s intracellular multiplication and evasion of the host immune response [113].
The gene expression profile of macrophages from C57BLl/6 and CBA mice was analyzed before and after infection by L. amazonensis and an increase in genes related to infection control in C57BLl/6 mice was reported, such as genes linked to apoptosis and phagocytosis, but not in CBA mice, where there was an increase in genes involved in lipid metabolism and, therefore, in the modulation of the parasitophorous vacuole [114].
Infection of peritoneal macrophages from C57BLl/6 mice with L. amazonensis and L. major demonstrated a non-generalized suppression of the lipopolysaccharide (LPS)-induced inflammatory response. It was observed that there was a reduction in the protein production of the cytokines IL-17, IL-12, IL-6, IL-13, and IL-3 induced by LPS, but an increase in TNF, IL-1α, MIP-1α, and MCP-1 [115]. In addition, the uptake of infected apoptotic cells induces the production of renders macrophages to produce transforming growth factor-β (TGF-β), creating an anti-inflammatory environment that promotes promoting L. major growth in macrophages [47]. Similar results were also presented with L. amazonensis by Afonso et al. [51].
More specifically, to survive within the hostile environment of macrophages, Leishmania has developed strategies to subvert the antimicrobial mechanisms mounted by these cells. These include the impairment of antigen presentation and cytokine secretion, sequestration of macrophage metabolic pathways, inhibition of NO, and induction of immunosuppressive molecules such as IL-10 and TGF-β; both cytokines involved in the deactivation of macrophage functions [116]. Furthermore, the induction of immunomodulatory molecules, such as CD200, by Leishmania also inhibits macrophage activation. In this same work, it was also shown that CD200-dependent inhibition of inducible nitric oxide synthase (iNOS) was responsible for the increased virulence of L. amazonensis [93,117].
In the context of the modulation of macrophage activation by Leishmania, studies in murine leishmaniasis models have shown that antibody production is associated with an unproductive immune response. Antibodies promote increased lesion size and parasite numbers during ineffective immunity towards L. major, L. mexicana, and L. amazonensis [88,118,119]. It was observed, in the study by MILES et al., that the presence of opsonized Leishmania seems to induce the production of IL-10, allowing the progression of the disease [88]. Furthermore, another study also demonstrated that the phagocytosis of apoptotic neutrophils by macrophages from C57BLl/6 mice leads to the induction of an M2b profile permissible for the L. major growth [120]. These data are reinforced by the observation of the relationship between the clinical phenotype of leishmaniasis and the magnitude of macrophage infection. Macrophages from individuals with chronic dermal leishmaniasis or recurrent disease were more permissive to Leishmania parasites than those from asymptomatically infected individuals [121,122]. These studies suggest that there may be an association between the response to Leishmania and the cell stage evaluated, with differentiated macrophages being more permissive to infection in vitro than monocytes [123]. In this sense, depending on the phenotype of monocytes/macrophages recruited to the site of infection and the infecting parasite species, these cells may contribute in different ways to host protection or disease immunopathology [124].

6. Leishmania and Oxidative Stress

The induction of cellular stress in Leishmania-infected macrophages is still a controversial topic. However, an increasing number of works indicate that Leishmania infection induces ROS production in macrophages [125,126] and several studies demonstrate the susceptibility of Leishmania to exogenous ROS and NO [4,127].
ROS are highly reactive oxygen-containing molecules, which can be free radicals (superoxide anion -O2-; hydroxyl radicals -OH; peroxynitrite -ONOO-, among others) or neutral molecules (hydrogen peroxide -H2O2; hypochlorous acid -HOCl; ozone -O3; singlet oxygen-1O2), among others [128]. ROS are known to activate various signal transduction pathways including the mitogen-activated protein kinases (MAPK), extracellular signal-regulated protein kinases 1 and 2 (ERK1/2), NF-κB, Nrf-2/Keap-1/ARE, and the PI3K/Akt pathway [129]. Several enzymes produce superoxide and other ROS, including the mitochondrial electron transport chain, NOS, cytochrome P450 oxidase, and xanthine oxidase. However, in all these systems, superoxide production occurs as a byproduct of other reactions. On the contrary, NADPH oxidase is the only enzyme whose primary function is the generation of superoxide/ROS and the main ROS-generating mechanism in macrophages [128].
Once produced, ROS can interact with a wide variety of biological molecules through electron donation, promoting cell signaling and regulation of numerous physiological processes including the regulation of immune cell functions and antitumor responses [130]. Several works described cell death because of NOX activation. Increased production of ROS is observed in cells that undergo apoptosis [131] and the use of exogenous ROS induces apoptosis in several cell types [132,133].
Upon recognition of Leishmania, properly activated macrophages become effector cells that can destroy phagocytosed pathogens through a variety of cellular processes including the production of lysosomal degradation enzymes, generation of oxidative stress, and production of NO [5,93]. Although Leishmania parasites are sensitive to ROS, the respiratory burst that occurs in non-activated macrophages following infection is insufficient to kill the parasites [134], which could be due to the parasites inhibiting ROS generation in phagolysosomes [135], even though both human and mouse monocytes produce high levels of ROS and can mediate ROS-dependent killing of Leishmania without prior activation [136,137]. Assreuy et al. demonstrated that the killing of L. major by IFNγ-activated murine macrophages in vitro was dependent on the production of NO, but not on the production of superoxide or peroxynitrite [138]. In humans, monocytes from patients with CL produced higher amounts of ROS after in vitro infection with L. (V.) braziliensis than healthy subjects. Moreover, the inhibition of ROS production in Leishmania-infected monocytes increased the presence of viable parasites, indicating their important role in parasite killing [139]. Gantt et al. demonstrated that both murine and human macrophages produce O2 and NO during phagocytosis of opsonized L. chagasi promastigotes and that both contribute to the intracellular death of the pathogen [125]. However, other studies demonstrate the opposite. Filardy et al. reported that L. major infection triggered a rapid cellular stress response, with increased production of ROS and proinflammatory signals, but is also involved in parasite survival and replication in host macrophages [38]. In addition, Blos et al. observed that Nox2-deficient animals infected with L. major controlled the infection in the initial moments, but they did not adequately control the replication of the parasite in the spleen, showing its importance in vivo at late moments [140].
The induction of suppressors of cytokine signaling (SOCS) proteins and thus apoptosis inhibition in L. donovani-infected macrophages was not affected by H2O2 treatment, which suggests the establishment of a replicative niche within the host [141]. Other studies also demonstrated the importance of ROS in the resistance to infection by L. guyanensis and L. amazonensis [142]. A possible role of superoxide and ONOO in the defense against L. amazonensis in vivo was also proposed [53,143,144].

7. Concluding Remarks

The importance of parasite–innate immune cell interactions became evident in both in vitro and in vivo models of Leishmania infection. We discussed the literature data on innate immune cells activity such as neutrophils, macrophages, and DCs, and the importance of their receptors and molecules in controlling Leishmania infection. In addition, we described different mechanisms that favor Leishmania survival and proliferation. Changes in signaling pathways and cell phenotype, as well as the increased production of a series of pro- and anti-inflammatory mediators, may contribute in a systematic way to the negative modulation of the immune response in the initial moments of infection. These mechanisms emerged because of a long parasite–host co-evolutionary process.
Additionally, some contradictory data found in the literature may reflect several variables including different infection models and the species of Leishmania used. Thus, the intimate connection between infection and the tissue repair response opens unexplored lines of research. Further investigation of innate immune cells and Leishmania can lead to a better understanding of the infectious process and to better vaccines for leishmaniasis.

Author Contributions

Conceptualization, A.C.C.-d.-S., D.d.O.N., C.G.F.-d.-L. and A.A.F.; writing, A.C.C.-d.-S., D.d.O.N., J.R.M.F., C.G.F.-d.-L., A.M., K.G.-P. and A.A.F.; supervision, C.G.F.-d.-L., A.A.F. and D.D.-R.; project administration, C.G.F.-d.-L., L.F.-d.-L. and A.A.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was supported by the Brazilian National Research Council (CNPq), Rio de Janeiro State Science Foundation (FAPERJ), Fundação Oswaldo Cruz (FIOCRUZ), and Programa Institutos Nacionais de Ciência e Tecnologia (INCT), CNPq, Brazil. We thank Lindomar Miranda for their helpful technical assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Choi, H.L.; Jain, S.; Ruiz Postigo, J.A.; Borisch, B.; Dagne, D.A. The global procurement landscape of leishmaniasis medicines. PLoS Negl. Trop. Dis. 2021, 15, e0009181. [Google Scholar] [CrossRef]
  2. Benallal, K.E.; Garni, R.; Harrat, Z.; Volf, P.; Dvorak, V. Phlebotomine sand flies (Diptera: Psychodidae) of the Maghreb region: A systematic review of distribution, morphology, and role in the transmission of the pathogens. PLoS Negl. Trop. Dis. 2022, 16, e0009952. [Google Scholar] [CrossRef]
  3. Daga, M.K.; Rohatgi, I.; Mishra, R. Leishmaniasis. Indian J. Crit. Care Med. 2021, 25, S166–S170. [Google Scholar] [CrossRef] [PubMed]
  4. Goto, H.; Lauletta Lindoso, J.A. Cutaneous and mucocutaneous leishmaniasis. Infect. Dis. Clin. N. Am. 2012, 26, 293–307. [Google Scholar] [CrossRef] [PubMed]
  5. Van Assche, T.; Deschacht, M.; da Luz, R.A.; Maes, L.; Cos, P. Leishmania-macrophage interactions: Insights into the redox biology. Free Radic. Biol. Med. 2011, 51, 337–351. [Google Scholar] [CrossRef] [PubMed]
  6. Mostafavi, E.; Ghasemian, A.; Abdinasir, A.; Nematollahi Mahani, S.A.; Rawaf, S.; Salehi Vaziri, M.; Gouya, M.M.; Minh Nhu Nguyen, T.; Al Awaidy, S.; Al Ariqi, L.; et al. Emerging and Re-emerging Infectious Diseases in the WHO Eastern Mediterranean Region, 2001–2018. Int. J. Health Policy Manag. 2021. [Google Scholar] [CrossRef] [PubMed]
  7. Mougneau, E.; Bihl, F.; Glaichenhaus, N. Cell biology and immunology of Leishmania. Immunol. Rev. 2011, 240, 286–296. [Google Scholar] [CrossRef] [PubMed]
  8. Serafim, T.D.; Coutinho-Abreu, I.V.; Dey, R.; Kissinger, R.; Valenzuela, J.G.; Oliveira, F.; Kamhawi, S. Leishmaniasis: The act of transmission. Trends Parasitol. 2021, 37, 976–987. [Google Scholar] [CrossRef]
  9. Mirzaei, A.; Maleki, M.; Masoumi, E.; Maspi, N. A historical review of the role of cytokines involved in leishmaniasis. Cytokine 2021, 145, 155297. [Google Scholar] [CrossRef] [PubMed]
  10. Blackwell, J.M.; Fakiola, M.; Castellucci, L.C. Human genetics of leishmania infections. Hum. Genet. 2020, 139, 813–819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Filardy, A.A.; Pires, D.R.; DosReis, G.A. Macrophages and neutrophils cooperate in immune responses to Leishmania infection. Cell Mol. Life Sci. 2011, 68, 1863–1870. [Google Scholar] [CrossRef] [PubMed]
  12. Arcanjo, A.F.; LaRocque-de-Freitas, I.F.; Rocha, J.D.; Zamith, D.; Costa-da-Silva, A.C.; Nunes, M.P.; Mesquita-Santos, F.P.; Morrot, A.; Filardy, A.A.; Mariano, M.; et al. The PGE2/IL-10 Axis Determines Susceptibility of B-1 Cell-Derived Phagocytes (B-1CDP) to Leishmania major Infection. PLoS ONE 2015, 10, e0124888. [Google Scholar] [CrossRef] [Green Version]
  13. Ehrchen, J.M.; Roth, J.; Roebrock, K.; Varga, G.; Domschke, W.; Newberry, R.; Sorg, C.; Müller-Tidow, C.; Sunderkötter, C.; Kucharzik, T.; et al. The absence of cutaneous lymph nodes results in a Th2 response and increased susceptibility to Leishmania major infection in mice. Infect. Immun. 2008, 76, 4241–4250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Kumar, R.; Bumb, R.A.; Salotra, P. Evaluation of localized and systemic immune responses in cutaneous leishmaniasis caused by Leishmania tropica: Interleukin-8, monocyte chemotactic protein-1 and nitric oxide are major regulatory factors. Immunology 2010, 130, 193–201. [Google Scholar] [CrossRef] [PubMed]
  15. Bacellar, O.; Lessa, H.; Schriefer, A.; Machado, P.; Ribeiro de Jesus, A.; Dutra, W.O.; Gollob, K.J.; Carvalho, E.M. Up-regulation of Th1-type responses in mucosal leishmaniasis patients. Infect. Immun. 2002, 70, 6734–6740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Hurdayal, R.; Nieuwenhuizen, N.E.; Revaz-Breton, M.; Smith, L.; Hoving, J.C.; Parihar, S.P.; Reizis, B.; Brombacher, F. Deletion of IL-4 receptor alpha on dendritic cells renders BALB/c mice hypersusceptible to Leishmania major infection. PLoS Pathog. 2013, 9, e1003699. [Google Scholar] [CrossRef] [Green Version]
  17. Anderson, C.F.; Stumhofer, J.S.; Hunter, C.A.; Sacks, D. IL-27 regulates IL-10 and IL-17 from CD4+ cells in nonhealing Leishmania major infection. J. Immunol. 2009, 183, 4619–4627. [Google Scholar] [CrossRef] [Green Version]
  18. Katara, G.K.; Raj, A.; Kumar, R.; Avishek, K.; Kaushal, H.; Ansari, N.A.; Bumb, R.A.; Salotra, P. Analysis of localized immune responses reveals presence of Th17 and Treg cells in cutaneous leishmaniasis due to Leishmania tropica. BMC Immunol. 2013, 14, 52. [Google Scholar] [CrossRef] [Green Version]
  19. Espir, T.T.; Figueira, L.E.P.; Naiff, M.E.F.; da Costa, A.G.; Ramalho-Ortigão, M.; Malheiro, A.; Franco, A.M. The role of inflammatory, anti-inflammatory, and regulatory cytokines in patients infected with cutaneous leishmaniasis in Amazonas State, Brazil. J. Immunol. Res. 2014, 2014, 481750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Dietze-Schwonberg, K.; Lopez Kostka, S.; Lorenz, B.; Regen, T.; Waisman, A.; von Stebut, E. IL-17A/F in Leishmania major-resistant C57BL/6 mice. Exp. Derm. 2019, 28, 321–323. [Google Scholar] [CrossRef]
  21. Gonçalves-de-Albuquerque, S.D.C.; Pessoa-E-Silva, R.; Trajano-Silva, L.A.M.; de Goes, T.C.; de Morais, R.C.S.; da C. Oliveira, C.N.; de Lorena, V.M.B.; de Paiva-Cavalcanti, M. The Equivocal Role of Th17 Cells and Neutrophils on Immunopathogenesis of Leishmaniasis. Front. Immunol. 2017, 8, 1437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Banerjee, A.; Bhattacharya, P.; Joshi, A.B.; Ismail, N.; Dey, R.; Nakhasi, H.L. Role of pro-inflammatory cytokine IL-17 in Leishmania pathogenesis and in protective immunity by Leishmania vaccines. Cell Immunol. 2016, 309, 37–41. [Google Scholar] [CrossRef] [PubMed]
  23. Von Stebut, E. Cutaneous Leishmania infection: Progress in pathogenesis research and experimental therapy. Exp. Derm. 2007, 16, 340–346. [Google Scholar] [CrossRef]
  24. Regli, I.B.; Passelli, K.; Hurrell, B.P.; Tacchini-Cottier, F. Survival Mechanisms Used by Some. Front. Immunol. 2017, 8, 1558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kolářová, I.; Valigurová, A. Hide-and-Seek: A Game Played between Parasitic Protists and Their Hosts. Microorganisms 2021, 9, 2434. [Google Scholar] [CrossRef] [PubMed]
  26. Filho, A.A.P.; Nascimento, A.A.S.; Saab, N.A.A.; Fugiwara, R.T.; D’Ávila Pessoa, G.C.; Koerich, L.B.; Pereira, M.H.; Araújo, R.N.; Sant’Anna, M.R.V.; Gontijo, N.F. Evasion of the complement system by Leishmania through the uptake of factor H, a complement regulatory protein. Acta Trop. 2021, 224, 106152. [Google Scholar] [CrossRef] [PubMed]
  27. Hermoso, T.; Fishelson, Z.; Becker, S.I.; Hirschberg, K.; Jaffe, C.L. Leishmanial protein kinases phosphorylate components of the complement system. EMBO J. 1991, 10, 4061–4067. [Google Scholar] [CrossRef] [PubMed]
  28. Nunes, A.C.; Almeida-Campos, F.R.; Horta, M.F.; Ramalho-Pinto, F.J. Leishmania amazonensis promastigotes evade complement killing by interfering with the late steps of the cascade. Parasitology 1997, 115 Pt 6, 601–609. [Google Scholar] [CrossRef]
  29. Domínguez, M.; Moreno, I.; Aizpurua, C.; Toraño, A. Early mechanisms of Leishmania infection in human blood. Microbes Infect. 2003, 5, 507–513. [Google Scholar] [CrossRef]
  30. Atayde, V.D.; Aslan, H.; Townsend, S.; Hassani, K.; Kamhawi, S.; Olivier, M. Exosome Secretion by the Parasitic Protozoan Leishmania within the Sand Fly Midgut. Cell Rep. 2015, 13, 957–967. [Google Scholar] [CrossRef] [Green Version]
  31. Dey, R.; Joshi, A.B.; Oliveira, F.; Pereira, L.; Guimarães-Costa, A.B.; Serafim, T.D.; de Castro, W.; Coutinho-Abreu, I.V.; Bhattacharya, P.; Townsend, S.; et al. Gut Microbes Egested during Bites of Infected Sand Flies Augment Severity of Leishmaniasis via Inflammasome-Derived IL-1β. Cell Host Microbe 2018, 23, 134–143.e136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Andrade, B.B.; de Oliveira, C.I.; Brodskyn, C.I.; Barral, A.; Barral-Netto, M. Role of sand fly saliva in human and experimental leishmaniasis: Current insights. Scand. J. Immunol. 2007, 66, 122–127. [Google Scholar] [CrossRef]
  33. Teixeira, M.J.; Teixeira, C.R.; Andrade, B.B.; Barral-Netto, M.; Barral, A. Chemokines in host-parasite interactions in leishmaniasis. Trends Parasitol. 2006, 22, 32–40. [Google Scholar] [CrossRef]
  34. Giraud, E.; Lestinova, T.; Derrick, T.; Martin, O.; Dillon, R.J.; Volf, P.; Műller, I.; Bates, P.A.; Rogers, M.E. Leishmania proteophosphoglycans regurgitated from infected sand flies accelerate dermal wound repair and exacerbate leishmaniasis via insulin-like growth factor 1-dependent signalling. PLoS Pathog. 2018, 14, e1006794. [Google Scholar] [CrossRef] [Green Version]
  35. Pacheco-Fernandez, T.; Volpedo, G.; Verma, C.; Satoskar, A.R. Understanding the immune responses involved in mediating protection or immunopathology during leishmaniasis. Biochem. Soc. Trans. 2021, 49, 297–311. [Google Scholar] [CrossRef] [PubMed]
  36. Goundry, A.; Romano, A.; Lima, A.P.C.A.; Mottram, J.C.; Myburgh, E. Inhibitor of serine peptidase 2 enhances Leishmania major survival in the skin through control of monocytes and monocyte-derived cells. FASEB J. 2018, 32, 1315–1327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Gwinn, M.R.; Vallyathan, V. Respiratory burst: Role in signal transduction in alveolar macrophages. J. Toxicol. Environ. Health B Crit. Rev. 2006, 9, 27–39. [Google Scholar] [CrossRef]
  38. Filardy, A.A.; Costa-da-Silva, A.C.; Koeller, C.M.; Guimarães-Pinto, K.; Ribeiro-Gomes, F.L.; Lopes, M.F.; Heise, N.; Freire-de-Lima, C.G.; Nunes, M.P.; DosReis, G.A. Infection with Leishmania major induces a cellular stress response in macrophages. PLoS ONE 2014, 9, e85715. [Google Scholar] [CrossRef] [PubMed]
  39. Hurrell, B.P.; Regli, I.B.; Tacchini-Cottier, F. Different Leishmania Species Drive Distinct Neutrophil Functions. Trends Parasitol. 2016, 32, 392–401. [Google Scholar] [CrossRef] [PubMed]
  40. Tacchini-Cottier, F.; Zweifel, C.; Belkaid, Y.; Mukankundiye, C.; Vasei, M.; Launois, P.; Milon, G.; Louis, J.A. An immunomodulatory function for neutrophils during the induction of a CD4+ Th2 response in BALB/c mice infected with Leishmania major. J. Immunol. 2000, 165, 2628–2636. [Google Scholar] [CrossRef] [Green Version]
  41. Charmoy, M.; Megnekou, R.; Allenbach, C.; Zweifel, C.; Perez, C.; Monnat, K.; Breton, M.; Ronet, C.; Launois, P.; Tacchini-Cottier, F. Leishmania major induces distinct neutrophil phenotypes in mice that are resistant or susceptible to infection. J. Leukoc. Biol. 2007, 82, 288–299. [Google Scholar] [CrossRef] [PubMed]
  42. Rochael, N.C.; Guimarães-Costa, A.B.; Nascimento, M.T.; DeSouza-Vieira, T.S.; Oliveira, M.P.; Garcia e Souza, L.F.; Oliveira, M.F.; Saraiva, E.M. Classical ROS-dependent and early/rapid ROS-independent release of Neutrophil Extracellular Traps triggered by Leishmania parasites. Sci. Rep. 2015, 5, 18302. [Google Scholar] [CrossRef] [Green Version]
  43. Moradin, N.; Descoteaux, A. Leishmania promastigotes: Building a safe niche within macrophages. Front. Cell. Infect. Microbiol. 2012, 2, 121. [Google Scholar] [CrossRef] [Green Version]
  44. Laufs, H.; Müller, K.; Fleischer, J.; Reiling, N.; Jahnke, N.; Jensenius, J.C.; Solbach, W.; Laskay, T. Intracellular survival of Leishmania major in neutrophil granulocytes after uptake in the absence of heat-labile serum factors. Infect. Immun. 2002, 70, 826–835. [Google Scholar] [CrossRef] [Green Version]
  45. Laskay, T.; van Zandbergen, G.; Solbach, W. Neutrophil granulocytes—Trojan horses for Leishmania major and other intracellular microbes? Trends Microbiol. 2003, 11, 210–214. [Google Scholar] [CrossRef]
  46. Charmoy, M.; Hurrell, B.P.; Romano, A.; Lee, S.H.; Ribeiro-Gomes, F.; Riteau, N.; Mayer-Barber, K.; Tacchini-Cottier, F.; Sacks, D.L. The Nlrp3 inflammasome, IL-1β, and neutrophil recruitment are required for susceptibility to a nonhealing strain of Leishmania major in C57BL/6 mice. Eur. J. Immunol. 2016, 46, 897–911. [Google Scholar] [CrossRef] [Green Version]
  47. van Zandbergen, G.; Klinger, M.; Mueller, A.; Dannenberg, S.; Gebert, A.; Solbach, W.; Laskay, T. Cutting edge: Neutrophil granulocyte serves as a vector for Leishmania entry into macrophages. J. Immunol. 2004, 173, 6521–6525. [Google Scholar] [CrossRef] [Green Version]
  48. Aga, E.; Katschinski, D.M.; van Zandbergen, G.; Laufs, H.; Hansen, B.; Müller, K.; Solbach, W.; Laskay, T. Inhibition of the spontaneous apoptosis of neutrophil granulocytes by the intracellular parasite Leishmania major. J. Immunol. 2002, 169, 898–905. [Google Scholar] [CrossRef] [Green Version]
  49. Peters, N.C.; Egen, J.G.; Secundino, N.; Debrabant, A.; Kimblin, N.; Kamhawi, S.; Lawyer, P.; Fay, M.P.; Germain, R.N.; Sacks, D. In vivo imaging reveals an essential role for neutrophils in leishmaniasis transmitted by sand flies. Science 2008, 321, 970–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Ribeiro-Gomes, F.L.; Otero, A.C.; Gomes, N.A.; Moniz-De-Souza, M.C.; Cysne-Finkelstein, L.; Arnholdt, A.C.; Calich, V.L.; Coutinho, S.G.; Lopes, M.F.; DosReis, G.A. Macrophage interactions with neutrophils regulate Leishmania major infection. J. Immunol. 2004, 172, 4454–4462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Afonso, L.; Borges, V.M.; Cruz, H.; Ribeiro-Gomes, F.L.; DosReis, G.A.; Dutra, A.N.; Clarêncio, J.; de Oliveira, C.I.; Barral, A.; Barral-Netto, M.; et al. Interactions with apoptotic but not with necrotic neutrophils increase parasite burden in human macrophages infected with Leishmania amazonensis. J. Leukoc. Biol. 2008, 84, 389–396. [Google Scholar] [CrossRef] [PubMed]
  52. Ribeiro-Gomes, F.L.; Moniz-de-Souza, M.C.; Alexandre-Moreira, M.S.; Dias, W.B.; Lopes, M.F.; Nunes, M.P.; Lungarella, G.; DosReis, G.A. Neutrophils activate macrophages for intracellular killing of Leishmania major through recruitment of TLR4 by neutrophil elastase. J. Immunol. 2007, 179, 3988–3994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Horta, M.F.; Mendes, B.P.; Roma, E.H.; Noronha, F.S.; Macêdo, J.P.; Oliveira, L.S.; Duarte, M.M.; Vieira, L.Q. Reactive oxygen species and nitric oxide in cutaneous leishmaniasis. J. Parasitol. Res. 2012, 2012, 203818. [Google Scholar] [CrossRef] [PubMed]
  54. Belkaid, Y.; Butcher, B.; Sacks, D.L. Analysis of cytokine production by inflammatory mouse macrophages at the single-cell level: Selective impairment of IL-12 induction in Leishmania-infected cells. Eur. J. Immunol. 1998, 28, 1389–1400. [Google Scholar] [CrossRef]
  55. Liew, F.Y.; Millott, S.; Parkinson, C.; Palmer, R.M.; Moncada, S. Macrophage killing of Leishmania parasite in vivo is mediated by nitric oxide from L-arginine. J. Immunol. 1990, 144, 4794–4797. [Google Scholar]
  56. Collin, M.; Bigley, V. Human dendritic cell subsets: An update. Immunology 2018, 154, 3–20. [Google Scholar] [CrossRef]
  57. Marovich, M.A.; McDowell, M.A.; Thomas, E.K.; Nutman, T.B. IL-12p70 production by Leishmania major-harboring human dendritic cells is a CD40/CD40 ligand-dependent process. J. Immunol. 2000, 164, 5858–5865. [Google Scholar] [CrossRef] [Green Version]
  58. Jafarzadeh, A.; Nemati, M.; Chauhan, P.; Patidar, A.; Sarkar, A.; Sharifi, I.; Saha, B. Interleukin-27 Functional Duality Balances. Front. Immunol. 2020, 11, 1573. [Google Scholar] [CrossRef]
  59. Moll, H.; Fuchs, H.; Blank, C.; Röllinghoff, M. Langerhans cells transport Leishmania major from the infected skin to the draining lymph node for presentation to antigen-specific T cells. Eur. J. Immunol. 1993, 23, 1595–1601. [Google Scholar] [CrossRef] [Green Version]
  60. Lemos, M.P.; Esquivel, F.; Scott, P.; Laufer, T.M. MHC class II expression restricted to CD8alpha+ and CD11b+ dendritic cells is sufficient for control of Leishmania major. J. Exp. Med. 2004, 199, 725–730. [Google Scholar] [CrossRef]
  61. Kautz-Neu, K.; Noordegraaf, M.; Dinges, S.; Bennett, C.L.; John, D.; Clausen, B.E.; von Stebut, E. Langerhans cells are negative regulators of the anti-Leishmania response. J. Exp. Med. 2011, 208, 885–891. [Google Scholar] [CrossRef] [Green Version]
  62. Ritter, U.; Meissner, A.; Scheidig, C.; Körner, H. CD8 alpha- and Langerin-negative dendritic cells, but not Langerhans cells, act as principal antigen-presenting cells in leishmaniasis. Eur. J. Immunol. 2004, 34, 1542–1550. [Google Scholar] [CrossRef] [PubMed]
  63. León, B.; López-Bravo, M.; Ardavín, C. Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania. Immunity 2007, 26, 519–531. [Google Scholar] [CrossRef] [Green Version]
  64. Biedermann, T.; Zimmermann, S.; Himmelrich, H.; Gumy, A.; Egeter, O.; Sakrauski, A.K.; Seegmüller, I.; Voigt, H.; Launois, P.; Levine, A.D.; et al. IL-4 instructs TH1 responses and resistance to Leishmania major in susceptible BALB/c mice. Nat. Immunol. 2001, 2, 1054–1060. [Google Scholar] [CrossRef]
  65. Hurdayal, R.; Nieuwenhuizen, N.E.; Khutlang, R.; Brombacher, F. Inflammatory Dendritic Cells, Regulated by IL-4 Receptor Alpha Signaling, Control Replication, and Dissemination of Leishmania major in mice. Front. Cell. Infect. Microbiol. 2019, 9, 479. [Google Scholar] [CrossRef]
  66. Bogdan, C. Natural killer cells in experimental and human leishmaniasis. Front. Cell. Infect. Microbiol. 2012, 2, 69. [Google Scholar] [CrossRef] [Green Version]
  67. Scharton, T.M.; 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. J. Exp. Med. 1993, 178, 567–577. [Google Scholar] [CrossRef] [PubMed]
  68. Messlinger, H.; Sebald, H.; Heger, L.; Dudziak, D.; Bogdan, C.; Schleicher, U. Monocyte-Derived Signals Activate Human Natural Killer Cells in Response to Leishmania Parasites. Front. Immunol. 2018, 9, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Naqvi, N.; Srivastava, R.; Selvapandiyan, A.; Puri, N. Host Mast Cells in Leishmaniasis: Friend or Foe? Trends Parasitol. 2020, 36, 952–956. [Google Scholar] [CrossRef]
  70. Maurer, M.; Lopez Kostka, S.; Siebenhaar, F.; Moelle, K.; Metz, M.; Knop, J.; von Stebut, E. Skin mast cells control T cell-dependent host defense in Leishmania major infections. FASEB J. 2006, 20, 2460–2467. [Google Scholar] [CrossRef]
  71. Lopez Kostka, S.; Dinges, S.; Griewank, K.; Iwakura, Y.; Udey, M.C.; von Stebut, E. IL-17 promotes progression of cutaneous leishmaniasis in susceptible mice. J. Immunol. 2009, 182, 3039–3046. [Google Scholar] [CrossRef]
  72. De Oliveira, M.P.; Lima, M.C.; Calheiros, A.S.; Martins, M.A.; Antas, P.R.; De Luca, P.M.; Pirmez, C. Leishmania (Viannia) braziliensis: Human mast cell line activation induced by logarithmic and stationary promastigote derived-lysates. Exp. Parasitol. 2005, 109, 72–79. [Google Scholar] [CrossRef]
  73. Wershil, B.K.; Theodos, C.M.; Galli, S.J.; Titus, R.G. Mast cells augment lesion size and persistence during experimental Leishmania major infection in the mouse. J. Immunol. 1994, 152, 4563–4571. [Google Scholar] [PubMed]
  74. Romão, P.R.; Da Costa Santiago, H.; Ramos, C.D.; De Oliveira, C.F.; Monteiro, M.C.; De Queiroz Cunha, F.; Vieira, L.Q. Mast cell degranulation contributes to susceptibility to Leishmania major. Parasite Immunol. 2009, 31, 140–146. [Google Scholar] [CrossRef]
  75. Ehrchen, J.M.; Roebrock, K.; Foell, D.; Nippe, N.; von Stebut, E.; Weiss, J.M.; Münck, N.A.; Viemann, D.; Varga, G.; Müller-Tidow, C.; et al. Keratinocytes determine Th1 immunity during early experimental leishmaniasis. PLoS Pathog. 2010, 6, e1000871. [Google Scholar] [CrossRef]
  76. Kane, M.M.; Mosser, D.M. Leishmania parasites and their ploys to disrupt macrophage activation. Curr. Opin. Hematol. 2000, 7, 26–31. [Google Scholar] [CrossRef] [PubMed]
  77. Aderem, A. Phagocytosis and the inflammatory response. J. Infect. Dis. 2003, 187 (Suppl. S2), S340–S345. [Google Scholar] [CrossRef] [PubMed]
  78. Mosser, D.M.; Vlassara, H.; Edelson, P.J.; Cerami, A. Leishmania promastigotes are recognized by the macrophage receptor for advanced glycosylation endproducts. J. Exp. Med. 1987, 165, 140–145. [Google Scholar] [CrossRef]
  79. Mosser, D.M.; Edelson, P.J. The third component of complement (C3) is responsible for the intracellular survival of Leishmania major. Nature 1987, 327, 329–331. [Google Scholar] [CrossRef] [PubMed]
  80. Wyler, D.J.; Sypek, J.P.; McDonald, J.A. In vitro parasite-monocyte interactions in human leishmaniasis: Possible role of fibronectin in parasite attachment. Infect. Immun. 1985, 49, 305–311. [Google Scholar] [CrossRef] [Green Version]
  81. Brittingham, A.; Chen, G.; McGwire, B.S.; Chang, K.P.; Mosser, D.M. Interaction of Leishmania gp63 with cellular receptors for fibronectin. Infect. Immun. 1999, 67, 4477–4484. [Google Scholar] [CrossRef] [Green Version]
  82. Vannier-Santos, M.A.; Saraiva, E.M.; Martiny, A.; Neves, A.; de Souza, W. Fibronectin shedding by Leishmania may influence the parasite-macrophage interaction. Eur. J. Cell Biol. 1992, 59, 389–397. [Google Scholar] [PubMed]
  83. Marth, T.; Kelsall, B.L. Regulation of interleukin-12 by complement receptor 3 signaling. J. Exp. Med. 1997, 185, 1987–1995. [Google Scholar] [CrossRef] [Green Version]
  84. Brittingham, A.; Mosser, D.M. Exploitation of the complement system by Leishmania promastigotes. Parasitol. Today 1996, 12, 444–447. [Google Scholar] [CrossRef]
  85. Belkaid, Y.; Hoffmann, K.F.; Mendez, S.; Kamhawi, S.; Udey, M.C.; Wynn, T.A.; Sacks, D.L. The role of interleukin (IL)-10 in the persistence of Leishmania major in the skin after healing and the therapeutic potential of anti-IL-10 receptor antibody for sterile cure. J. Exp. Med. 2001, 194, 1497–1506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Belkaid, Y.; Piccirillo, C.A.; Mendez, S.; Shevach, E.M.; Sacks, D.L. CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity. Nature 2002, 420, 502–507. [Google Scholar] [CrossRef]
  87. Kane, M.M.; Mosser, D.M. The role of IL-10 in promoting disease progression in leishmaniasis. J. Immunol. 2001, 166, 1141–1147. [Google Scholar] [CrossRef] [Green Version]
  88. Miles, S.A.; Conrad, S.M.; Alves, R.G.; Jeronimo, S.M.; Mosser, D.M. A role for IgG immune complexes during infection with the intracellular pathogen Leishmania. J. Exp. Med. 2005, 201, 747–754. [Google Scholar] [CrossRef]
  89. Kima, P.E.; Constant, S.L.; Hannum, L.; Colmenares, M.; Lee, K.S.; Haberman, A.M.; Shlomchik, M.J.; McMahon-Pratt, D. Internalization of Leishmania mexicana complex amastigotes via the Fc receptor is required to sustain infection in murine cutaneous leishmaniasis. J. Exp. Med. 2000, 191, 1063–1068. [Google Scholar] [CrossRef] [Green Version]
  90. Von Stebut, E.; Belkaid, Y.; Jakob, T.; Sacks, D.L.; Udey, M.C. Uptake of Leishmania major amastigotes results in activation and interleukin 12 release from murine skin-derived dendritic cells: Implications for the initiation of anti-Leishmania immunity. J. Exp. Med. 1998, 188, 1547–1552. [Google Scholar] [CrossRef] [Green Version]
  91. Woelbing, F.; Kostka, S.L.; Moelle, K.; Belkaid, Y.; Sunderkoetter, C.; Verbeek, S.; Waisman, A.; Nigg, A.P.; Knop, J.; Udey, M.C.; et al. Uptake of Leishmania major by dendritic cells is mediated by Fcgamma receptors and facilitates acquisition of protective immunity. J. Exp. Med. 2006, 203, 177–188. [Google Scholar] [CrossRef] [Green Version]
  92. Ueno, N.; Wilson, M.E. Receptor-mediated phagocytosis of Leishmania: Implications for intracellular survival. Trends Parasitol. 2012, 28, 335–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Liu, D.; Uzonna, J.E. The early interaction of Leishmania with macrophages and dendritic cells and its influence on the host immune response. Front. Cell. Infect. Microbiol. 2012, 2, 83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Faria, M.S.; Reis, F.C.; Lima, A.P. Toll-like receptors in leishmania infections: Guardians or promoters? J. Parasitol. Res. 2012, 2012, 930257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Gallego, C.; Golenbock, D.; Gomez, M.A.; Saravia, N.G. Toll-like receptors participate in macrophage activation and intracellular control of Leishmania (Viannia) panamensis. Infect. Immun. 2011, 79, 2871–2879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Komai-Koma, M.; Li, D.; Wang, E.; Vaughan, D.; Xu, D. Anti-Toll-like receptor 2 and 4 antibodies suppress inflammatory response in mice. Immunology 2014, 143, 354–362. [Google Scholar] [CrossRef] [Green Version]
  97. Vargas-Inchaustegui, D.A.; Tai, W.; Xin, L.; Hogg, A.E.; Corry, D.B.; Soong, L. Distinct roles for MyD88 and Toll-like receptor 2 during Leishmania braziliensis infection in mice. Infect. Immun. 2009, 77, 2948–2956. [Google Scholar] [CrossRef] [Green Version]
  98. Carneiro, P.P.; Dórea, A.S.; Oliveira, W.N.; Guimarães, L.H.; Brodskyn, C.; Carvalho, E.M.; Bacellar, O. Blockade of TLR2 and TLR4 Attenuates Inflammatory Response and Parasite Load in Cutaneous Leishmaniasis. Front. Immunol. 2021, 12, 706510. [Google Scholar] [CrossRef]
  99. Abou Fakher, F.H.; Rachinel, N.; Klimczak, M.; Louis, J.; Doyen, N. TLR9-dependent activation of dendritic cells by DNA from Leishmania major favors Th1 cell development and the resolution of lesions. J. Immunol. 2009, 182, 1386–1396. [Google Scholar] [CrossRef] [Green Version]
  100. Liese, J.; Schleicher, U.; Bogdan, C. TLR9 signaling is essential for the innate NK cell response in murine cutaneous leishmaniasis. Eur. J. Immunol. 2007, 37, 3424–3434. [Google Scholar] [CrossRef]
  101. Walker, D.M.; Oghumu, S.; Gupta, G.; McGwire, B.S.; Drew, M.E.; Satoskar, A.R. Mechanisms of cellular invasion by intracellular parasites. Cell Mol. Life Sci. 2014, 71, 1245–1263. [Google Scholar] [CrossRef] [Green Version]
  102. McConville, M.J.; de Souza, D.; Saunders, E.; Likic, V.A.; Naderer, T. Living in a phagolysosome; metabolism of Leishmania amastigotes. Trends Parasitol. 2007, 23, 368–375. [Google Scholar] [CrossRef] [PubMed]
  103. Chaudhuri, G.; Chaudhuri, M.; Pan, A.; Chang, K.P. Surface acid proteinase (gp63) of Leishmania mexicana. A metalloenzyme capable of protecting liposome-encapsulated proteins from phagolysosomal degradation by macrophages. J. Biol. Chem. 1989, 264, 7483–7489. [Google Scholar] [CrossRef]
  104. Isnard, A.; Shio, M.T.; Olivier, M. Impact of Leishmania metalloprotease GP63 on macrophage signaling. Front. Cell. Infect. Microbiol. 2012, 2, 72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Rodriguez, N.E.; Chang, H.K.; Wilson, M.E. Novel program of macrophage gene expression induced by phagocytosis of Leishmania chagasi. Infect. Immun. 2004, 72, 2111–2122. [Google Scholar] [CrossRef] [Green Version]
  106. Pandey, S.P.; Doyen, N.; Mishra, G.C.; Saha, B.; Chandel, H.S. TLR9-deficiency reduces TLR1, TLR2 and TLR3 expressions in Leishmania major-infected macrophages. Exp. Parasitol. 2015, 154, 82–86. [Google Scholar] [CrossRef]
  107. Buates, S.; Matlashewski, G. General suppression of macrophage gene expression during Leishmania donovani infection. J. Immunol. 2001, 166, 3416–3422. [Google Scholar] [CrossRef] [Green Version]
  108. Dillon, L.A.; Okrah, K.; Hughitt, V.K.; Suresh, R.; Li, Y.; Fernandes, M.C.; Belew, A.T.; Corrada Bravo, H.; Mosser, D.M.; El-Sayed, N.M. Transcriptomic profiling of gene expression and RNA processing during Leishmania major differentiation. Nucleic Acids Res. 2015, 43, 6799–6813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Kalavi, K.; Jorjani, O.; Faghihi, M.A.; Mowla, S.J. Cytokine Gene Expression Alterations in Human Macrophages Infected by. Cell J. 2021, 22, 476–481. [Google Scholar] [CrossRef] [PubMed]
  110. Chaussabel, D.; Semnani, R.T.; McDowell, M.A.; Sacks, D.; Sher, A.; Nutman, T.B. Unique gene expression profiles of human macrophages and dendritic cells to phylogenetically distinct parasites. Blood 2003, 102, 672–681. [Google Scholar] [CrossRef] [PubMed]
  111. Shadab, M.; Das, S.; Banerjee, A.; Sinha, R.; Asad, M.; Kamran, M.; Maji, M.; Jha, B.; Deepthi, M.; Kumar, M.; et al. RNA-Seq revealed expression of many novel genes associated with Leishmania donovani persistence and clearance in the host macrophage. Front. Cell. Infect. Microbiol. 2019, 9, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Osorio y Fortéa, J.; de La Llave, E.; Regnault, B.; Coppée, J.Y.; Milon, G.; Lang, T.; Prina, E. Transcriptional signatures of BALB/c mouse macrophages housing multiplying Leishmania amazonensis amastigotes. BMC Genom. 2009, 10, 119. [Google Scholar] [CrossRef] [Green Version]
  113. Orikaza, C.M.; Pessoa, C.C.; Paladino, F.V.; Florentino, P.T.V.; Barbiéri, C.L.; Goto, H.; Ramos-Sanchez, E.M.; Franco da Silveira, J.; Rabinovitch, M.; Mortara, R.A.; et al. Dual Host-Intracellular Parasite Transcriptome of Enucleated Cells Hosting Leishmania amazonensis: Control of Half-Life of Host Cell Transcripts by the Parasite. Infect. Immun. 2020, 88, e00261-20. [Google Scholar] [CrossRef] [PubMed]
  114. Probst, C.M.; Silva, R.A.; Menezes, J.P.; Almeida, T.F.; Gomes, I.N.; Dallabona, A.C.; Ozaki, L.S.; Buck, G.A.; Pavoni, D.P.; Krieger, M.A.; et al. A comparison of two distinct murine macrophage gene expression profiles in response to Leishmania amazonensis infection. BMC Microbiol. 2012, 12, 22. [Google Scholar] [CrossRef] [Green Version]
  115. Lapara, N.J.; Kelly, B.L. Suppression of LPS-induced inflammatory responses in macrophages infected with Leishmania. J. Inflamm. 2010, 7, 8. [Google Scholar] [CrossRef] [Green Version]
  116. Arango Duque, G.; Descoteaux, A. Leishmania survival in the macrophage: Where the ends justify the means. Curr. Opin. Microbiol. 2015, 26, 32–40. [Google Scholar] [CrossRef] [PubMed]
  117. Cortez, M.; Huynh, C.; Fernandes, M.C.; Kennedy, K.A.; Aderem, A.; Andrews, N.W. Leishmania promotes its own virulence by inducing expression of the host immune inhibitory ligand CD200. Cell Host Microbe 2011, 9, 463–471. [Google Scholar] [CrossRef] [Green Version]
  118. Buxbaum, L.U. A detrimental role for IgG and FcgammaR in Leishmania mexicana infection. Immunol. Res. 2008, 42, 197–209. [Google Scholar] [CrossRef]
  119. Wanasen, N.; Xin, L.; Soong, L. Pathogenic role of B cells and antibodies in murine Leishmania amazonensis infection. Int. J. Parasitol. 2008, 38, 417–429. [Google Scholar] [CrossRef] [Green Version]
  120. Filardy, A.A.; Pires, D.R.; Nunes, M.P.; Takiya, C.M.; Freire-de-Lima, C.G.; Ribeiro-Gomes, F.L.; DosReis, G.A. Proinflammatory clearance of apoptotic neutrophils induces an IL-12(low)IL-10(high) regulatory phenotype in macrophages. J. Immunol. 2010, 185, 2044–2050. [Google Scholar] [CrossRef] [Green Version]
  121. Bosque, F.; Saravia, N.G.; Valderrama, L.; Milon, G. Distinct innate and acquired immune responses to Leishmania in putative susceptible and resistant human populations endemically exposed to L. (Viannia) panamensis infection. Scand. J. Immunol. 2000, 51, 533–541. [Google Scholar] [CrossRef] [Green Version]
  122. Robledo, S.; Wozencraft, A.; Valencia, A.Z.; Saravia, N. Human monocyte infection by Leishmania (Viannia) panamensis. Role of complement receptors and correlation of susceptibility in vitro with clinical phenotype. J. Immunol. 1994, 152, 1265–1276. [Google Scholar]
  123. Bosque, F.; Milon, G.; Valderrama, L.; Saravia, N.G. Permissiveness of human monocytes and monocyte-derived macrophages to infection by promastigotes of Leishmania (Viannia) panamensis. J. Parasitol. 1998, 84, 1250–1256. [Google Scholar] [CrossRef] [PubMed]
  124. Loría-Cervera, E.N.; Andrade-Narvaez, F. The role of monocytes/macrophages in Leishmania infection: A glance at the human response. Acta Trop. 2020, 207, 105456. [Google Scholar] [CrossRef] [PubMed]
  125. Gantt, K.R.; Goldman, T.L.; McCormick, M.L.; Miller, M.A.; Jeronimo, S.M.; Nascimento, E.T.; Britigan, B.E.; Wilson, M.E. Oxidative responses of human and murine macrophages during phagocytosis of Leishmania chagasi. J. Immunol. 2001, 167, 893–901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Deschacht, M.; Van Assche, T.; Hendrickx, S.; Bult, H.; Maes, L.; Cos, P. Role of oxidative stress and apoptosis in the cellular response of murine macrophages upon Leishmania infection. Parasitology 2012, 139, 1429–1437. [Google Scholar] [CrossRef]
  127. Furtado, R.R.; Soares, D.C.; Prado, A.F.; Farias, L.H.S.; Da Silva, B.J.M.; Rodrigues, A.P.D.; Silva, E.O. Constitutive nitric oxide synthase-like enzyme in two species involved in cutaneous and mucocutaneous leishmaniasis. Parasitol. Int. 2021, 83, 102347. [Google Scholar] [CrossRef]
  128. Coso, S.; Harrison, I.; Harrison, C.B.; Vinh, A.; Sobey, C.G.; Drummond, G.R.; Williams, E.D.; Selemidis, S. NADPH oxidases as regulators of tumor angiogenesis: Current and emerging concepts. Antioxid. Redox Signal. 2012, 16, 1229–1247. [Google Scholar] [CrossRef] [Green Version]
  129. Zhang, Z.; Wang, L.; Du, J.; Li, Y.; Yang, H.; Li, C.; Li, H.; Hu, H. Lipid raft localization of epidermal growth factor receptor alters matrix metalloproteinase-1 expression in SiHa cells via the MAPK/ERK signaling pathway. Oncol. Lett. 2016, 12, 4991–4998. [Google Scholar] [CrossRef] [Green Version]
  130. Sena, L.A.; Chandel, N.S. Physiological roles of mitochondrial reactive oxygen species. Mol. Cell 2012, 48, 158–167. [Google Scholar] [CrossRef] [Green Version]
  131. Alexandre, J.; Batteux, F.; Nicco, C.; Chéreau, C.; Laurent, A.; Guillevin, L.; Weill, B.; Goldwasser, F. Accumulation of hydrogen peroxide is an early and crucial step for paclitaxel-induced cancer cell death both in vitro and in vivo. Int. J. Cancer 2006, 119, 41–48. [Google Scholar] [CrossRef] [PubMed]
  132. Conde de la Rosa, L.; Schoemaker, M.H.; Vrenken, T.E.; Buist-Homan, M.; Havinga, R.; Jansen, P.L.; Moshage, H. Superoxide anions and hydrogen peroxide induce hepatocyte death by different mechanisms: Involvement of JNK and ERK MAP kinases. J. Hepatol. 2006, 44, 918–929. [Google Scholar] [CrossRef] [PubMed]
  133. Rayner, B.S.; Duong, T.T.; Myers, S.J.; Witting, P.K. Protective effect of a synthetic anti-oxidant on neuronal cell apoptosis resulting from experimental hypoxia re-oxygenation injury. J. Neurochem. 2006, 97, 211–221. [Google Scholar] [CrossRef] [PubMed]
  134. Nacy, C.A.; Groves, M.G. Macrophages in resistance to rickettsial infections: Early host defense mechanisms in experimental scrub typhus. Infect. Immun. 1981, 31, 1239–1250. [Google Scholar] [CrossRef] [Green Version]
  135. Matheoud, D.; Moradin, N.; Bellemare-Pelletier, A.; Shio, M.T.; Hong, W.J.; Olivier, M.; Gagnon, E.; Desjardins, M.; Descoteaux, A. Leishmania evades host immunity by inhibiting antigen cross-presentation through direct cleavage of the SNARE VAMP8. Cell Host Microbe 2013, 14, 15–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Goncalves, R.; Zhang, X.; Cohen, H.; Debrabant, A.; Mosser, D.M. Platelet activation attracts a subpopulation of effector monocytes to sites of Leishmania major infection. J. Exp. Med. 2011, 208, 1253–1265. [Google Scholar] [CrossRef]
  137. Novais, F.O.; Nguyen, B.T.; Beiting, D.P.; Carvalho, L.P.; Glennie, N.D.; Passos, S.; Carvalho, E.M.; Scott, P. Human classical monocytes control the intracellular stage of Leishmania braziliensis by reactive oxygen species. J. Infect. Dis. 2014, 209, 1288–1296. [Google Scholar] [CrossRef]
  138. Assreuy, J.; Cunha, F.Q.; Epperlein, M.; Noronha-Dutra, A.; O’Donnell, C.A.; Liew, F.Y.; Moncada, S. Production of nitric oxide and superoxide by activated macrophages and killing of Leishmania major. Eur. J. Immunol. 1994, 24, 672–676. [Google Scholar] [CrossRef]
  139. Carneiro, P.P.; Conceição, J.; Macedo, M.; Magalhães, V.; Carvalho, E.M.; Bacellar, O. The Role of Nitric Oxide and Reactive Oxygen Species in the Killing of Leishmania braziliensis by Monocytes from Patients with Cutaneous Leishmaniasis. PLoS ONE 2016, 11, e0148084. [Google Scholar] [CrossRef] [Green Version]
  140. Blos, M.; Schleicher, U.; Soares Rocha, F.J.; Meissner, U.; Röllinghoff, M.; Bogdan, C. Organ-specific and stage-dependent control of Leishmania major infection by inducible nitric oxide synthase and phagocyte NADPH oxidase. Eur. J. Immunol. 2003, 33, 1224–1234. [Google Scholar] [CrossRef]
  141. Srivastav, S.; Basu Ball, W.; Gupta, P.; Giri, J.; Ukil, A.; Das, P.K. Leishmania donovani prevents oxidative burst-mediated apoptosis of host macrophages through selective induction of suppressors of cytokine signaling (SOCS) proteins. J. Biol. Chem. 2014, 289, 1092–1105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Sousa-Franco, J.; Araújo-Mendes, E.; Silva-Jardim, I.; L-Santos, J.; Faria, D.R.; Dutra, W.O.; Horta, M.F. Infection-induced respiratory burst in BALB/c macrophages kills Leishmania guyanensis amastigotes through apoptosis: Possible involvement in resistance to cutaneous leishmaniasis. Microbes Infect. 2006, 8, 390–400. [Google Scholar] [CrossRef]
  143. Linares, E.; Giorgio, S.; Augusto, O. Inhibition of in vivo leishmanicidal mechanisms by tempol: Nitric oxide down-regulation and oxidant scavenging. Free Radic. Biol. Med. 2008, 44, 1668–1676. [Google Scholar] [CrossRef]
  144. Mukbel, R.M.; Patten, C.; Gibson, K.; Ghosh, M.; Petersen, C.; Jones, D.E. Macrophage killing of Leishmania amazonensis amastigotes requires both nitric oxide and superoxide. Am. J. Trop. Med. Hyg. 2007, 76, 669–675. [Google Scholar] [CrossRef]
Figure 1. Impact of Leishmania infection on immune cells. Leishmania spp. interacts with multiple innate immune cells modulating their phenotype and function, as well as the adaptive immune responses. Mast cells collaborate in disease progression by secreting IL-4 and IL-13 fostering Th2 responses and parasite survival (panel 1). Neutrophils, macrophages, and DCs can either eliminate or promote parasite survival. Recruited neutrophils eliminate leishmanial parasites through phagocytosis, ROS, and NETs release. Leishmania can survive transiently within neutrophils by inhibiting phagolysosome biogenesis and oxidative stress, and by delaying neutrophil apoptosis. Infected neutrophils also secrete IL-8 and MIP1β, which attract additional neutrophils and other phagocytic cells, favoring Leishmania survival and pathology (panel 2). Macrophages can be differentiated in M1 or M2 during leishmaniasis. M1 macrophages produce proinflammatory cytokine and chemokines, NO and ROS, booster Th1 responses, and favor disease control. M2 macrophages increase the production of IL-10 and TGFβ, and support Th2 response and disease progression (panel 3). DCs regulate immune responses against Leishmania by migrating to the draining lymph nodes to present Leishmania-derived antigen to naïve T cells. DCs can induce the differentiation of Th1 by secreting IL-12 and IL-27 or Th2, by blocking IL-12 secretion (panel 4). NK cells have a protective role in leishmaniasis by secreting IFNγ to boost Th1 response (panel 5). IL; interleukin IFNγ; ROS; reactive oxidative species, NO; nitric oxide, NETs; neutrophil extracellular traps, MIP; macrophage; inflammatory protein, TNFα; tumor necrosis factor α, TGFβ; transforming growth factor-β, NK; natural killer.
Figure 1. Impact of Leishmania infection on immune cells. Leishmania spp. interacts with multiple innate immune cells modulating their phenotype and function, as well as the adaptive immune responses. Mast cells collaborate in disease progression by secreting IL-4 and IL-13 fostering Th2 responses and parasite survival (panel 1). Neutrophils, macrophages, and DCs can either eliminate or promote parasite survival. Recruited neutrophils eliminate leishmanial parasites through phagocytosis, ROS, and NETs release. Leishmania can survive transiently within neutrophils by inhibiting phagolysosome biogenesis and oxidative stress, and by delaying neutrophil apoptosis. Infected neutrophils also secrete IL-8 and MIP1β, which attract additional neutrophils and other phagocytic cells, favoring Leishmania survival and pathology (panel 2). Macrophages can be differentiated in M1 or M2 during leishmaniasis. M1 macrophages produce proinflammatory cytokine and chemokines, NO and ROS, booster Th1 responses, and favor disease control. M2 macrophages increase the production of IL-10 and TGFβ, and support Th2 response and disease progression (panel 3). DCs regulate immune responses against Leishmania by migrating to the draining lymph nodes to present Leishmania-derived antigen to naïve T cells. DCs can induce the differentiation of Th1 by secreting IL-12 and IL-27 or Th2, by blocking IL-12 secretion (panel 4). NK cells have a protective role in leishmaniasis by secreting IFNγ to boost Th1 response (panel 5). IL; interleukin IFNγ; ROS; reactive oxidative species, NO; nitric oxide, NETs; neutrophil extracellular traps, MIP; macrophage; inflammatory protein, TNFα; tumor necrosis factor α, TGFβ; transforming growth factor-β, NK; natural killer.
Tropicalmed 07 00054 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Costa-da-Silva, A.C.; Nascimento, D.d.O.; Ferreira, J.R.M.; Guimarães-Pinto, K.; Freire-de-Lima, L.; Morrot, A.; Decote-Ricardo, D.; Filardy, A.A.; Freire-de-Lima, C.G. Immune Responses in Leishmaniasis: An Overview. Trop. Med. Infect. Dis. 2022, 7, 54. https://doi.org/10.3390/tropicalmed7040054

AMA Style

Costa-da-Silva AC, Nascimento DdO, Ferreira JRM, Guimarães-Pinto K, Freire-de-Lima L, Morrot A, Decote-Ricardo D, Filardy AA, Freire-de-Lima CG. Immune Responses in Leishmaniasis: An Overview. Tropical Medicine and Infectious Disease. 2022; 7(4):54. https://doi.org/10.3390/tropicalmed7040054

Chicago/Turabian Style

Costa-da-Silva, Ana Caroline, Danielle de Oliveira Nascimento, Jesuino R. M. Ferreira, Kamila Guimarães-Pinto, Leonardo Freire-de-Lima, Alexandre Morrot, Debora Decote-Ricardo, Alessandra Almeida Filardy, and Celio Geraldo Freire-de-Lima. 2022. "Immune Responses in Leishmaniasis: An Overview" Tropical Medicine and Infectious Disease 7, no. 4: 54. https://doi.org/10.3390/tropicalmed7040054

Article Metrics

Back to TopTop