Next Article in Journal
HIV Latency in Myeloid Cells: Challenges for a Cure
Next Article in Special Issue
Eosinophils, but Not Type 2 Innate Lymphoid Cells, Are the Predominant Source of Interleukin 4 during the Innate Phase of Leishmania major Infection
Previous Article in Journal
New Insights in Acanthamoeba
Previous Article in Special Issue
Hemoglobin Endocytosis and Intracellular Trafficking: A Novel Way of Heme Acquisition by Leishmania
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Visceral Leishmaniasis and the Skin: Dermal Parasite Transmission to Sand Flies

1
Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA
2
Immunology Program, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
3
Center for Emerging Infectious Diseases, University of Iowa Research Park, Coralville, IA 52241, USA
*
Author to whom correspondence should be addressed.
Submission received: 10 March 2022 / Revised: 12 May 2022 / Accepted: 19 May 2022 / Published: 24 May 2022
(This article belongs to the Special Issue Leishmania & Leishmaniasis)

Abstract

:
Visceral leishmaniasis is a parasitic disease with significant dermal tropism. The skin is an important site of infection contributing to parasite transmission to naïve sand flies, but understanding how parasitism of host skin and the related immune microenvironment supports or prevents skin parasite replication is now the focus of major investigation in the field of leishmaniasis research. Here, we review dermatoimmunology during visceral leishmaniasis (VL), dermal Leishmania parasite burden, and the role of skin parasitism in transmissibility to sand fly vectors. First, we discuss the epidemiology of VL amongst dogs, the primary zoonotic reservoir for human infection. We explore the association between spatial distribution and the burden of parasites in the skin in driving outward transmission. Factors associated with parasite persistence in the skin are examined. We discuss systemic immunity during VL and what is known about immunological correlates in the skin microenvironment. Finally, we touch on factors egested into the skin during Leishmania inoculation by sand flies. Throughout, we discuss factors associated with the early and chronic establishment of Leishmania parasites in the skin and the role of the dermal immune response.

1. Introduction

Visceral leishmaniasis (VL) is caused by Leishmania donovani complex trypanosomatid protozoan parasites. It is a fatal disease in humans with >90% case fatality within two years if left undetected and untreated [1]. VL due to L. donovani spp. is a zoonotic disease that causes 50,000 to 90,000 new human cases, more than 50,000 deaths, and countless canine cases per year [2,3]. This review seeks to explore recent evidence regarding the dermotropic nature of visceralizing species of Leishmania, the dermal immune environment during VL, and the role of dermal parasite burden in outward transmission to naïve sand flies.
We utilized the PubMed database to compile data from primary manuscripts and review articles regarding visceral leishmaniasis species and the skin occurring over the last two decades. To our knowledge, this is the first review of this subject. This literature review was carried out using the PubMed database. Key search words used included “leishmaniasis”, “xenodiagnoses”, “infectivity”, “transmission”, “immunology”, “skin”, and “dermal”. Original articles were prioritized over case reports or conference proceedings, and articles were initially chosen from the last 20 years except where seminal papers from prior years were used or suggested by reviewers.
As new evidence emerges regarding how skin parasite burden predicts infectiousness from host to vector, we believed it was necessary to delve further into the literature on the implications of the dermal immune environment’s role in skin parasite burden and subsequent outward transmission. It is important to better understand the factors affecting outward transmission from infected hosts, as this is a key component of the transmission cycle for preventive interventions to be targeted. Elucidation of the immunological mechanisms that exacerbate dermal parasite accumulation may have implications for the skin’s potential role in outward transmission. This could also mean the skin is a prime target for immunological interventions, such as topical biologics, in breaking the parasite transmission cycle.
The review details the epidemiology of VL and the importance of canid reservoirs in transmission. We then go on to discuss how both the burden and spatial distribution of dermal parasite burden are related to outward transmission to vectors. We then examine the causes of parasite persistence in bite sites and the contribution parasite persistence has to the buildup of parasite burden in the skin. We first look at the early and then the chronic establishment of dermal parasite burden from an immunological perspective. Lastly, we consider the immune influence of components transferred from sand flies into the skin during bites and the subsequent inflammation from a sand fly bite in the skin. Our goal is to review the literature regarding multiple factors that contribute to dermal parasite burden and emphasize its role in outward transmission.

2. Epidemiology

Leishmania infantum is the primary causative strain of VL across the Mediterranean basin (Southern Europe, North Africa, the Middle East) and the Americas [4]. Leishmania donovani is the primary cause of VL in East Africa and South Asia [5]. As of 2020, the bulk of VL cases occurred amongst impoverished, vulnerable communities, with 90% of cases occurring in just 10 countries: India, Sudan, South Sudan, Ethiopia, China, Eritrea, Kenya, Somalia, Yemen, and Brazil [6]. L. donovani complex spp. are also known to occasionally cause cutaneous leishmaniasis (CL) in Sri Lanka. L. infantum is known to cause CL in northern India, northern Pakistan, northern Iran, and Turkey. [7,8]. L. donovani is currently considered anthroponotic, although a role for animal reservoir hosts is possible [9,10], and L. infantum resides in canine reservoirs in the Americas as well as the Mediterranean basin. L. infantum was imported to North America via hunting dogs from Southern Europe [11], and L. infantum within the North American hunting hound population is enzootic [12,13,14]. Using data collected over 15 years, Toepp et al. calculated an incidence rate of 25 cases of VL per 1000 dogs [15]. Regions of the Mediterranean such as Italy have reported CanL incidence rates as high as 9.5% [16]. Surveillance studies of dogs in endemic areas of Brazil put prevalence as high as 22% by PCR testing [17]. Parasites from vertically infected North American hounds are still infectious to naïve competent sand flies in an experimental setting [18]. Three species of Leishmania competent sand fly vectors are thought to be present in North America: Lutzomyia anthophora, Lu. diabolica, and Lu. shannoni [19,20]. Sand fly vector distribution patterns in Europe have changed in the past few decades primarily due to climate change and other environmental factors. Changes in vector distribution patterns in North America may similarly occur due to environmental changes [21,22]. Although Lu. shannoni in North America has not been proven to be a leishmaniasis vector according to the modified Killick-Kendrick criteria for incriminating natural vectors [23], it has been shown to possibly be a vector for L. infantum in Brazil [24], and changes in vector distribution may potentially allow North American sand fly species to fulfill the vector incrimination criteria. This could potentially pose a zoonotic risk of outward domestic transmission from naturally infected canine reservoirs to proximate human populations, particularly in the Southwest, South, and Southeastern U.S., where sand flies are endemic [19,20]. Canine reservoir status and similarities between human and canine L. infantum-specific immunology make dogs a public health-relevant animal model for studying VL pathogenesis, intervention, and therapeutic strategies.

3. Transmission and Dermal Parasite Burden

Leishmania parasites are transmitted between mammalian hosts via female phlebotomine sand flies and vertical, transplacental infection [25,26,27]. Although VL is primarily known to affect organs such as the bone marrow, liver, and spleen, there is evidence to suggest parasitism of the skin in most infected mammals. Dermal parasite burden may accumulate due to the skin being the initial inoculation site and maintained through multiple immune mechanisms, including a unique inflammatory environment in the skin, regulatory cell proliferation, protein induction, macrophage recruitment, T cell exhaustion, and parasite immune evasion factors. The dermotropic nature of Leishmania has been shown to be critical for outward transmission to sand flies, with skin parasite burden being a predominating factor for infectiousness [18,28].
L. infantum has been shown to be significantly dermotropic, and skin parasite burden is highly correlated with parasite transmission to sand flies, even in congenitally infected dogs [18,28]. Skin parasite burden correlates with parasitemia, with skin parasite burden consistently higher than blood parasite burden [18]. Parasitemia, therefore, can be comparatively lower and is not always the best predictor of outward transmission [18,29]. Splenic parasite burden, generally thought to be one of the most parasitized tissues in VL, was comparable to skin parasite burden [18,28]. One study in dogs reported significant positive correlations between parasite load in skin and sand fly infection rate and sand fly parasite load post xenodiagnoses [30].
Although there was a correlation between the dermal parasite burden of dogs and transmission to sand flies, it is not clear if there is a definitive correlation between disease severity and transmission [28]. Laurenti et al. showed that the clinical severity of dogs infected with L. infantum was inversely correlated with the infection rate of sand flies in a xenodiagnosis study; all but one asymptomatic dog was able to transmit parasites to a naïve vector while a lower proportion of symptomatic dogs was able to do so [31]. Another study found that the infection rate of naïve sand flies fed on infected dogs had no correlation with the symptoms or clinical group of the dog [32]. Despite these findings, several other studies have found significant positive correlations between clinical severity and infectiousness. Courtenay et al. found that seropositive dogs in Brazil undergoing xenodiagnosis exhibited a positive correlation between clinical disease and infectiousness, and only 17% of the most highly infectious dogs accounted for >80% of sand fly infections [33]. A similar positive correlation between the clinical severity of dogs and infectiousness to sand flies experimentally has been reported in several other xenodiagnoses studies in Brazil [34,35,36,37,38] and one study in Colombia [39]. A study in the U.S. showed that dogs with mild or moderate VL had the highest transmissibility from the skin to sand fly vector, while severe disease did not [18]. Another xenodiagnoses study investigating ear skin, specifically from Brazilian dogs, found that relative numbers of L. infantum in ear skin increased with the duration and severity of infection [28]. A meta-analysis of data published up until 2009 found that the proportion of infectious dogs is significantly positively correlated with clinical severity [40]. Xenodiagnoses studies in other mammalian models such as hamsters showed that L. donovani transmission from sick hamsters to sand flies was surprisingly low, but new flies fed on the same site acquired significantly more infections [41]. In L. major infected BALB/c mice, repeated sand fly bites increased the parasite loads in the skin but did not alter infectiousness [42]. Taken together, infectiousness to sand flies was most associated with high skin parasite burden, while disease state and parasitemia were not as predictive of infectiousness.
Experiments investigating outward transmission from human skin to naïve sand flies, emphasizing the role of skin in transmissibility, have also been conducted. In humans, xenodiagnoses studies in VL patients due to L. donovani in Bihar, India, have shown that 55% of patients with VL were infectious to sand flies before treatment. Modeling using this data demonstrated that every 10-fold increase in skin qPCR parasite load increased the odds of infecting a naïve sand fly 1.0–5.53 times [43]. A study from Ethiopia used skin microbiopsies to detect L. donovani parasites. The study reported that microbiopsies had a 10-fold smaller volume of blood than finger stick samples yet yielded higher Leishmania DNA rates, indicating the skin may be a more accessible source of parasites for diagnostics compared with blood [44]. Volunteers with a history of VL were also just as likely as healthy volunteers to test positive for L. donovani on microbiopsy, meaning there are many more asymptomatic cases than previously thought [44]. In each endemic location, even small numbers of infectious dogs or humans have the capacity to successfully infect a proportionate amount of sand flies with parasites. While it is known that L. donovani parasites persist in human skin, it remains to be definitively confirmed whether infected human skin itself promotes L. donovani transmission to sand flies. Xenodiagnoses studies of HIV patients coinfected with L. infantum have been performed, and these studies found that a significant proportion of coinfected patients were highly infectious to sand flies [45,46]. One xenodiagnoses study conducted in Spain found two immunocompetent VL patients with the active but untreated disease were able to transmit L. infantum to sand flies [47]. The studies discussed above are part of a growing body of evidence highlighting the potential infectious capability of VL-infected human skin to sand fly vectors in an experimental setting.
Post-kala-azar dermal leishmaniasis (PKDL) is a serious cutaneous manifestation of VL that can occur after VL drug treatment presenting as nodular/papular or macular lesions [48]. Risk factors are believed to be immunosuppression or an immune reaction after VL drug treatment [48]. Patients with PKDL can infect naïve sand flies, illustrating a further example of outward transmission from the skin [49,50]. Over 50% of PKDL patients were capable of transmitting parasites to naïve sand flies. Of the PKDL patients that did demonstrate outward transmission, a higher skin parasite load by a factor of 10 was observed when compared with PDKL patients that were incapable of outward transmission [51]. Further studies investigating nonlesional skin of both symptomatic and asymptomatic VL patients are needed to further elucidate the potential of infected human skin for outward transmission and whether exposed areas are more likely to transmit parasites due to particular aspects of skin parasite distribution.

4. Dermal Distribution of Leishmania and Outward Transmission

L. donovani spp. have been shown to distribute heterogeneously in the skin of RAG-deficient mice lacking T and B cells after infection, creating heterogeneous pools of parasites in the skin that correlate with infectiousness and transmissibility [29,52]. The larger and denser a parasite pool is, the more likely transmission from that animal to a sand fly. The patchy distribution of parasites in the skin is an important finding because although patchiness decreases the expected number of sand flies acquiring parasites, it increases the infection load of a sand fly when it does find a particular patch, increasing transmissibility. Reservoir animals such as dogs can be “super-spreaders”, in which a small percentage of dogs harbored almost 90% of total parasites [28]. If a small percentage of reservoirs not only harbor the majority of parasites but distribute them in the skin in such a heterogenous way as to increase transmissibility, those dogs may be especially infectious to sand flies, posing an increased threat to human and animal health in endemic locations. Multiple blood feedings can also increase the frequency of parasite transmission [53]. The patchiness of Leishmania parasite burden in the skin has been shown to be true in an immunosuppressed (RAG1 deficient) mouse model, but it has not yet been shown to be true of L. infantum in a canine model. We speculate that based on the dermotropic nature of L. infantum and patchiness in a mouse model, the skin of infected dogs would also have heterogeneous, patchy distribution of parasites. This skin parasite landscape will be important for prediction models of infectiousness of other important hosts, dogs, or people. Modeling skin parasite residence is significant when sampling skin because it might explain confounding factors that affect host infectiousness and outward transmission. Doehl et al. used an endogenously fluorescent parasite model intravenously injected into a RAG-deficient mouse and combined data with spatial point pattern analysis to identify several model mechanisms to determine why the skin parasite landscape was patchy [54]. Their model suggested that parasites were initially seeded into the skin before uptake by myeloid cells [54]. Parasitized myeloid cells then aggregated in patches similar to granulomas. Uninfected myeloid cells recruited to cellular aggregates were infected by parasites from the initial seeding patch, creating self-propagating clusters of patches. Parasite patch size was highly variable, but patch centers tended to be more densely parasitized than the periphery, increasing the likelihood of parasite uptake from the center of patches [54]. Higher parasite loads accompanied by a more intense inflammatory infiltrate have been reported in the ear, ungual, and ventral abdomen regions of infected dogs [36], but other studies report no preferred skin site of parasite distribution on a micro level [54]. Skin wounds and trauma have been shown to recruit monocytes to the deep dermal layer as early as neutrophils infiltration [55]. Patients with Hansen’s disease (leprosy) caused by Mycobacterium leprae experience severe dermal lesions and inflammation due to infection, with numerous manifestations of dermal pathology. Hansen’s disease-associated dermal reactions can be misdiagnosed as PKDL, as they are similar to a dermal lesion [56]. It may be that the inflammatory environment in the skin during comorbidities that create lesional skin predisposes the skin to both infection via vector and parasite patch development due to the overwhelming presence of macrophages at lesional sites in the skin prior to Leishmania infection. We speculate that the initial bite serves as skin trauma that not only recruits monocytes but also sets up a future patch location. This is especially important for animals that live in groups, such as hunting dogs, whose faces and ears are often traumatized by other dogs, setting up areas of high monocyte infiltration that can then become an initial seeding parasite patch that self-propagates via clustering during chronic infection. Whether parasites prefer proximity to vasculature for uptake of nutrients is unclear, skin parasite distribution in relation to vasculature needs to be studied more closely. The spatial distribution and density of parasite distribution in the skin of infected mammals is emerging as a critical factor in outward transmission to sand flies.

5. Parasite Persistence at Bite Sites

The sand fly is a telmophage; its proboscis lacerates the skin of the host, causing localized damage in the dermis and creating a blood pool on which to feed. This allows for parasite uptake from both dermal blood vessels and dermal macrophages with phagocytosed parasites [29]. Similar to patches that occur in areas of trauma, there has been a specific study of parasite persistence in the skin at bite sites. Parasite persistence is as important to consider as spatial organization of parasites for outward transmission and belie a temporal component to parasite accumulation and density of parasitic patches in the skin. After the early stages of infection, parasites persist in patches for months to years, while chronic infection develops with consequent immune modulation of parasite-controlling mechanisms. L. infantum parasites persisted for at least six months in ulcerative skin lesions at primary bite sites, but parasites were also observed to persist for six months in normal-looking skin at a secondary bite site [57]. Sand flies were also found to acquire parasites after feeding on lesions at the primary bite site [57]. These findings allude to the ability of parasitized yet noninflamed or lesional skin to continue to be infectious to sand flies. Parasites can build up in the skin over time, even in clinically healthy mammals, and those parasites can still be outwardly infectious to sand flies.

6. Systemic Immunity and Correlations to the Skin

The skin immune environment contributes to preventing inflammation and allowing the buildup of skin parasite patches. The immune environment transitions from an inflammatory to a regulatory environment as chronic infection is established, contributing to parasite accumulation in the skin (Figure 1). It has long been established that high levels of interleukin-10 (IL-10) in keratinocytes are a strong predictor of the development of post-kala-azar dermal leishmaniasis in patients treated for visceral leishmaniasis [58]. There is also a higher expression of programmed death-ligand 1 (PD-L1) and indoleamine 2,3-dioxygenase (IDO1) checkpoint molecules on CD68+ monocytes and macrophages infected with parasites compared with noninfected monocytes and macrophages [59].
After treatment, patients with L. donovani cutaneous leishmaniasis expressed reduced PD-L1 and IDO1 in macrophages [59]. An early reduction of PD-L1 expression predicted a faster rate of clinical cure in parallel with a reduction in parasite load [59]. Symptomatic L. infantum-infected dogs demonstrate a four-fold increase or two-fold increase in PD-1 surface expression on CD4+ T cells and CD8+ T cells, respectively, compared with healthy controls [38]. Blockage of PD-L1 with anti-PD-L1 antibody partially restored CD4+ and CD8+ T cell proliferation, CD4+ interferon-gamma (IFNy) production, and increased macrophage reactive oxygen species (ROS) production, altogether resulting in decreased macrophage parasite burden [60]. CD4+ T cell effector responses may also be important for preventing infectiousness of host animals; Guarga et al. found that L. infantum-infected dogs with lower CD4+ T cell counts had higher rates of infectiousness to the vector [61]. This supports a role for PD-L1 checkpoint inhibitor drugs used either topically or systemically as part of an antileishmanial regimen or PD-L1 expression as a predictor of clinical outcomes [59,60]. This also supports the idea of a regulatory environment in the skin being more conducive to parasite patch coalescence.
B cells are associated with PD-L1 and the creation of a regulatory environment as clinical VL progresses [60,62]. A cytosolic tryparedoxin from L. infantum specifically activated B cells and caused them to secrete IL-10 and immunoglobulin G (IgG) [63]. Regulatory IgDhi B cells drive IL-10 production in progressive VL dogs from Brazil [62]. This illustrates the importance of IL-10 for establishing chronic, latent infection. Blockage of IL-10 receptor has shown success in achieving a sterile cure in chronically infected mice but did not reach the same level of efficacy in studies of human or dog cells [64]. Studies on PD-L1 and IL-10 receptor inhibitors together suggest potential therapeutic avenues to pursue a sterile cure in chronically infected individuals with VL. PD-L1, B cells, and IL-10 all contribute to a regulatory skin environment and the establishment of chronic dermal Leishmania infection.
Immune cell and cytokine profiles of the skin can show clear delineations between infected and uninfected dogs. Immunohistochemistry studies of VL skin have demonstrated more cells of all types, particularly CD8+ T cells and macrophages, and increased IFNy and inducible nitric oxide synthase (iNOS) production in the dermis of lesioned skin from patients with L. infantum nonulcerated cutaneous leishmaniasis compared with the skin of healthy individuals [65]. Experimental studies looking at immune gene expression in leishmanin skin test positive reactions of Ibizan hounds saw significant upregulation of Toll-like receptor 2 (TLR2), IL10, IFNy, and PDL1 compared with the healthy skin of endemic control hounds [66]. Toll-like receptor 7 (TLR7) was significantly downregulated [66]. A prevalence of CD3+ and large mononuclear cells, as opposed to B cells, coupled with a high expression of TLR2 indicate an ongoing delayed-type hypersensitivity response during these skin test reactions [66]. Upregulation of these TLR genes also aligned with a competent immune response in these dogs and support a role for TLR2 as protective against Leishmania. In lesional skin from L. infantum-infected dogs, there were increased neutrophils, histiocytes, T cells, and GATA3+, and IL-17a+ cells compared with the skin from uninfected dogs [67]. Normal skin from infected dogs also had more histiocytes, T cells, and GATA3+ compared with uninfected dogs but was also more abundant in FoxP3+ cells [67]. These findings support the role of dermal IFNy-mediated inflammation in protective immunity. It is interesting to note the upregulation of FoxP3+ cells in normal skin from uninfected dogs since studies in L. infantum-infected murine spleen and lymph nodes have shown high levels of CD4+ CD25+ regulatory T cells and FoxP3 expression that may contribute to immunosuppression of effector responses and subsequent control of immunopathology [68]. It is possible that a similar mechanism is responsible for immunosuppression in the skin and protection from lesions in chronically infected dogs and people. Lesions and ulcers can also become infected with bacteria, leading to inflammation that can be protective against Leishmania in the skin [69].

7. Sand Fly Saliva, Neutrophils, and Macrophages in Skin

As previously described, the presence of macrophages in the skin can contribute to both parasite accumulation and burden. Most of what is known about initial events after Leishmania infection is from cutaneous L. major mouse experimental infections. In this setting, there was rapid and sustained neutrophil infiltration at localized sand fly bites from which neutrophils captured Leishmania parasites [70]. Neutrophils were attracted to the wound by interleukin-1B (IL-1β), CXCL1, and yellow salivary proteins of the sand fly [71]. Sand flies egest part of their gut microbiome with every bite, which triggers inflammasome production of IL-1β, sustaining neutrophil recruitment and infiltration at the bite site [72]. Sand fly salivary proteins contribute to sustained immune cell recruitment and the establishment of initial infection and dermal parasite burden. Lymphocyte antigen 6 complex locus G6D (Ly6G+) neutrophils rapidly invade the dermis after parasite inoculation, followed by macrophages, natural killer (NK) cells, and then B cells [73]. In addition to sand fly microbiota and saliva components, infected sand fly bites also deliver parasite-produced components promastigote secretory gel (PSG) and Leishmania exosomes into the skin [74,75]. PSG is a powerful macrophage recruitment molecule in the skin and enhances the synthesis of polyamines necessary for intracellular parasite growth [75]. Leishmania exosomes are vesicles with a proinflammatory capacity and the ability to recruit neutrophils to the bite site, exacerbating the influx of innate immune cells that can be infected and harbor parasites [74]. Neutrophils create an epidermal block within 1 hour of a sand fly bite [70]. It might be expected that neutrophils aid in parasite clearance; however, depletion of neutrophils at the bite site actually reduces the number of viable parasites in the skin, one of many Leishmania immune evasion strategies [70]. Parasite burden then shifts to macrophage populations, without immune activation, via phagocytic uptake after 6–7 days, according to a model that has been dubbed the “Trojan Horse theory” [70]. The mannose receptor is part of the C-type lectin family with the ability to bind and internalize many different endogenous and pathogen-derived ligands for antigen processing and presentation [76]. In Leishmania-resistant C57BL/6 mice, L. major produces nonhealing cutaneous lesions where parasites can be taken up by mannose receptor (MR)-positive dermal macrophages exhibiting M2 characteristics despite systemic Th1 inflammatory states [77]. M2 macrophages are alternatively activated by exposure to cytokines such as IL-4, IL-10, or IL-13 and are associated with wound healing and tissue repair via proliferation induction [78]. This MRhigh dermal macrophage population is maintained near the lesion by IL-4 and IL-10 [77]. Newer studies indicate that dermis-resident macrophages are the predominant phagocyte to engulf L. major in a murine model in the first 24 h of transmission by sand fly bite [79]. Confocal imaging shows infected neutrophils transferring their phagocytosed parasites to tissue-resident macrophages (TRMs) in situ, and TRMs engulf apoptotic-infected neutrophils in the skin in vitro [79]. RNA transcriptomic data from lesions in patients with CL demonstrated significantly higher expression of PD-L1 on parasite-infected monocytes and macrophages [59]. Less is known from VL patients, but a high number of M1 macrophages were detected in skin biopsies from patients infected with L. infantum, which also causes nonulcerated CL [80]. Cytosolic tryparedoxin secreted by L. infantum is one of many immune evasion pathways that helps the parasite combat ROS in macrophages, contributing to parasite resistance to the innate immune system [63]. Sand fly salivary proteins acted as chemoattractants leading to subsequent recruitment of macrophages for parasitism at the bite site, which allowed parasites to build up in the skin, completing the early stage of infection. During natural infection in dogs, L. infantum parasites produced increased RNA transcripts of metalloproteases in the skin, particularly at ear edges [81]. These proteases may serve as virulence factors contributing to dermal parasite burden and dermal tropism.

8. Sand Fly Bites and Skin

Events early in infection directly after the sand fly bite contribute to the dermal immune environment altering parasite persistence and transmissibility. When sand flies damage blood vessels, erythrocytes leak into tissue and are phagocytosed by macrophages, leading to the production of heme-oxygenase-1 (HO-1) [82]. HO-1 is also induced by sand fly saliva at bite sites early in the infection process in a mouse model [83]. Carbon monoxide is a major end product of HO-1 enzymatic reactions and suppresses inflammation in the skin and in infected cells, increasing parasite survival [83]. Inhibition of HO-1 enhances inflammation and subsequent tissue damage [82]. HO-1 from erythrocytes was induced by sand fly saliva to be an immune evasion tactic by Leishmania, as shown in a murine model. By suppressing inflammation in the skin, parasites thrived, contributing to parasite burden in mice.
Along with HO-1 induction, sand fly inflammatory salivary antigens elicited localized inflammation in murine models [57,71,72,84]. Sand fly salivary components are known to be vasodilating and immunomodulatory. Adenosine and AMP found in Phlebotomus species saliva had both properties [85]. Maxadilan from Lutzomyia sand fly species was a potent vasodilator [86]. Antimaxadilan vaccination protected mice from L. major infection [87]. Sand fly saliva has immunomodulatory effects on murine T cells, dendritic cells, and neutrophils [88]. Dogs immunized with 2 of 35 sand fly salivary proteins developed a strong cellular immune response at the bite site, characterized by increased IFNy and IL-12 production [84]. Although sand fly saliva is generally anti-inflammatory, resulting in less inflammation during parasite inoculation, prior immunization against parasites causes saliva-induced local inflammation at a nascent bite site, resulting in an adaptive immune response to saliva that can escalate to severe allergic responses and anaphylaxis. Sand fly saliva components are currently being considered for vaccine candidates because of their immunomodulatory properties. Similar to HO-1 enzyme, sand fly saliva suppresses inflammation in the skin, potentially allowing for parasites to evade immune destruction and thrive.
Keratinocytes are the primary epithelial cell in the skin and make up much of the dermal cell repertoire. Keratinocyte interplay with immune cells and damage to keratinocytes can set up the immune milieu occurring at the skin interface at the time of parasite exposure. Exposure of human keratinocytes to different species of Leishmania elicits distinct pro- or anti-inflammatory responses. The epidermis is a major source of cytokine expression that can determine the transition from a protective Th1 inflammatory response to a regulatory Th2 switch resulting in disease progression [89]. Mice resistant to L. major infection demonstrate significant gene production of IL-12, IL-1B, and IL-4 in the epidermis, promoting Th1 differentiation and resistance during the time of Th1/2 differentiation [89]. Supporting this, other experiments using murine keratinocytes incubated with L. infantum demonstrated upregulation of inflammatory cytokine genes for IL-6, IL-8, TNF-a, and IL-1b [90]. In a murine model, parasites accumulate in phagocytic cells in the reticular dermis more than in the epidermis or hypodermis [54]. These studies exemplify the role of keratinocytes in eliciting a proinflammatory response at the bite site and initiating disease. Keratinocyte contribution to a proinflammatory environment has an effect on parasite persistence in the skin and subsequent outward transmission of parasites.

9. Summary

VL is an important neglected tropical disease with a significant global burden. Understanding what effects transmission is crucial for control of the disease. VL is unequivocally dermotropic even in visceralizing species, and skin parasite burden best predicts transmission. Parasite-driven events and dermal immune mechanisms control parasite skin burden. Sand fly salivary antigens and HO-1 induction promote both inflammation and immunosuppression, as well as the recruitment of favored host cells—macrophages—to the bite site. Parasitism of recruited macrophages and propagation into parasite patches establishes parasite persistence in the skin. Expression of PD-L1 and IL-10 secreted by regulatory B cells contribute to the transition from a proinflammatory to regulatory environment systemically, which is likely to directly impact the establishment of chronic dermal infection. The immunology of the skin, in turn, influences skin parasite burden. Densely parasitized patches of the skin provide an ideal transmission situation for sand flies from both human patients and naturally infected canine reservoirs.

10. Conclusions

Further examination of the dermal immunology of the skin as it pertains to Leishmania infection is important for understanding the transmission of visceralizing Leishmania spp. Further elucidation of immunological mechanisms involved in outward transmission from the skin to sand flies will inform the creation of topical immunomodulatory therapeutics that can prevent outward transmission. Topical PD-L1 inhibitors or IL-1 receptor antagonists could decrease dermal inflammation of infected hosts and prevent vector transmission from infected dogs or people if specific cytokines are found to be associated with increased infectiousness. Establishment of L. infantum persistence in the skin and the dermal immune environment of humans infected with VL must be further explored. Understanding parasite trafficking to the skin and immune regulation of skin parasite burdens is critical for intervening in transmission and curbing the infection cycle for One Health.

Author Contributions

Writing—original draft preparation, S.A.; writing—review and editing, B.M.S. and C.P. 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.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. Kala-Azar Elimination Programme: Report of a WHO Consultation of Partners; WHO: Geneva, Switzerland, 10–11 February 2015. Available online: https://apps.who.int/iris/handle/10665/185042 (accessed on 16 November 2021).
  2. Kaye, P.M.; Aebischer, T. Visceral leishmaniasis: Immunology and prospects for a vaccine. Clin. Microbiol. Infect. 2011, 17, 1462–1470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Vida, B.; Toepp, A.; Schaut, R.G.; Esch, K.J.; Juelsgaard, R.; Shimak, R.M.; Petersen, C.A. Immunologic progression of canine leishmaniosis following vertical transmission in United States dogs. Vet. Immunol. Immunopathol. 2015, 169, 34–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Maurício, I.; Stothard, R.; Miles, M. The Strange Case of Leishmania chagasi. Parasitol. Today 2000, 16, 188–189. [Google Scholar] [CrossRef]
  5. Chappuis, F.; Sundar, S.; Hailu, A.; Ghalib, H.; Rijal, S.; Peeling, R.W.; Alvar, J.; Boelaert, M. Visceral leishmaniasis: What are the needs for diagnosis, treatment and control? Nat. Rev. Microbiol. 2007, 5, 873–882. [Google Scholar] [CrossRef] [PubMed]
  6. World Health Organization. Leishmaniasis. Available online: https://www.who.int/health-topics/leishmaniasis#tab=tab_1 (accessed on 1 May 2022).
  7. Siriwardana, H.V.Y.D.; Noyes, H.A.; Beeching, N.J.; Chance, M.L.; Karunaweera, N.D.; Bates, P.A. Leishmania donovaniand Cutaneous Leishmaniasis, Sri Lanka. Emerg. Infect. Dis. 2007, 13, 476–478. [Google Scholar] [CrossRef]
  8. Thakur, L.; Singh, K.K.; Shanker, V.; Negi, A.; Jain, A.; Matlashewski, G.; Jain, M. Atypical leishmaniasis: A global perspective with emphasis on the Indian subcontinent. PLoS Negl. Trop. Dis. 2018, 12, e0006659. [Google Scholar] [CrossRef]
  9. Singh, N.; Mishra, J.; Singh, R.; Singh, S. Animal Reservoirs of Visceral Leishmaniasis in India. J. Parasitol. 2013, 99, 64–67. [Google Scholar] [CrossRef]
  10. Kushwaha, A.K.; Scorza, B.M.; Singh, O.P.; Rowton, E.; Lawyer, P.; Sundar, S.; Petersen, C.A. Domestic mammals as reservoirs for Leishmania donovani on the Indian subcontinent: Possibility and consequences on elimination. Transbound. Emerg. Dis. 2022, 69, 268–277. [Google Scholar] [CrossRef]
  11. Franssen, S.; Bennett, C.; Petersen, C.A.; Cotton, J. Origin and Genetic Comparison of Vertically Transmitted Leishmania Leishmania Infantum from United States Dogs. Emerg. Infect. Dis. 2019, in press.
  12. Espinosa, O.A.; Serrano, M.G.; Camargo, E.P.; Teixeira, M.M.G.; Shaw, J.J. An appraisal of the taxonomy and nomenclature of trypanosomatids presently classified as Leishmania and Endotrypanum. Parasitology 2016, 145, 430–442. [Google Scholar] [CrossRef]
  13. Marcili, A.; Sperança, M.A.; da Costa, A.P.; Madeira, M.D.F.; Soares, H.S.; Sanches, C.D.O.; Acosta, I.D.C.; Girotto, A.; Minervino, A.H.; Horta, M.C.; et al. Phylogenetic relationships of Leishmania species based on trypanosomatid barcode (SSU rDNA) and gGAPDH genes: Taxonomic revision of Leishmania (L.) infantum chagasi in South America. Infect. Genet. Evol. 2014, 25, 44–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Schantz, P.M.; Steurer, F.J.; Duprey, Z.H.; Kurpel, K.P.; Barr, S.C.; Jackson, J.E.; Breitschwerdt, E.B.; Levy, M.G.; Fox, J.C. Autochthonous visceral leishmaniasis in dogs in North America. J. Am. Vet. Med. Assoc. 2005, 226, 1316–1322. [Google Scholar] [CrossRef] [PubMed]
  15. Toepp, A.J.; Schaut, R.G.; Scott, B.D.; Mathur, D.; Berens, A.J.; Petersen, C.A. Leishmania incidence and prevalence in U.S. hunting hounds maintained via vertical transmission. Vet. Parasitol. Reg. Stud. Rep. 2017, 10, 75–81. [Google Scholar] [CrossRef]
  16. Paradies, P.; Capelli, G.; Cafarchia, C.; De Caprariis, D.; Sasanelli, M.; Otranto, M. Incidences of Canine Leishmaniasis in an Endemic Area of Southern Italy. J. Vet. Med. Ser. B 2006, 53, 295–298. [Google Scholar] [CrossRef] [PubMed]
  17. Oliveira, M.R.; Neto, M.B.O.; Bezerra, T.L.; da Silva, W.S.I.; da Paz, W.S.; dos Santos, I.G.; Bezerra-Santos, M.; Lima, V.F.S. Canine leishmaniasis in an endemic region, Northeastern Brazil: A comparative study with four groups of animals. Parasitol. Res. 2021, 120, 3915–3923. [Google Scholar] [CrossRef] [PubMed]
  18. Scorza, B.M.; Mahachi, K.G.; Cox, A.C.; Toepp, A.J.; Leal-Lima, A.; Kushwaha, A.K.; Kelly, P.; Meneses, C.; Wilson, G.; Gibson-Corley, K.N.; et al. Leishmania infantum xenodiagnosis from vertically infected dogs reveals significant skin tropism. PLoS Negl. Trop. Dis. 2021, 15, e0009366. [Google Scholar] [CrossRef] [PubMed]
  19. Schaut, R.G.; Robles-Murguia, M.; Juelsgaard, R.; Esch, K.J.; Bartholomay, L.C.; Ramalho-Ortigao, M.; Petersen, C.A. Vectorborne Transmission ofLeishmania infantumfrom Hounds, United States. Emerg. Infect. Dis. 2015, 21, 2209–2212. [Google Scholar] [CrossRef] [Green Version]
  20. Young, D.; Perkins, P. Phlebotomine Sand Flies of North America (Diptera: Psychodidae). In Mosquito News; American Mosquito Control Association: Mount Laurel, NJ, USA, 1984; Volume 44, pp. 263–304. [Google Scholar]
  21. Chalghaf, B.; Chemkhi, J.; Mayala, B.; Harrabi, M.; Benie, G.B.; Michael, E.; Ben Salah, A. Ecological niche modeling predicting the potential distribution of Leishmania vectors in the Mediterranean basin: Impact of climate change. Parasites Vectors 2018, 11, 461. [Google Scholar] [CrossRef]
  22. Koch, L.K.; Kochmann, J.; Klimpel, S.; Cunze, S. Modeling the climatic suitability of leishmaniasis vector species in Europe. Sci. Rep. 2017, 7, 13325. [Google Scholar] [CrossRef]
  23. Ready, P.D. Biology of Phlebotomine Sand Flies as Vectors of Disease Agents. Annu. Rev. Èntomol. 2013, 58, 227–250. [Google Scholar] [CrossRef]
  24. Travi, B.; Ferro, C.; Cadena, H.; Montoya-Lerma, J.; Adler, G. Canine visceral leishmaniasis: Dog infectivity to sand flies from non-endemic areas. Res. Vet. Sci. 2002, 72, 83–86. [Google Scholar] [CrossRef] [PubMed]
  25. Grinnage-Pulley, T.; Scott, B.; Petersen, C.A. A Mother’s Gift: Congenital Transmission of Trypanosoma and Leishmania Species. PLoS Pathog. 2016, 12, e1005302. [Google Scholar] [CrossRef] [PubMed]
  26. Mescouto-Borges, M.R.M.; Maués, É.; Costa, D.L.; Pranchevicius, M.-C.S.; Romero, G. Congenitally transmitted visceral leishmaniasis: Report of two Brazilian human cases. Braz. J. Infect. Dis. 2013, 17, 263–266. [Google Scholar] [CrossRef] [Green Version]
  27. Osorio, Y.; Bonilla, D.L.; Henao, H.; Rodriguez, L.D.; Travi, B.L.; Peniche, A.; Saldarriaga, O. Congenital Transmission of Experimental Leishmaniasis in a Hamster Model. Am. J. Trop. Med. Hyg. 2012, 86, 812–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Courtenay, O.; Carson, C.; Calvo-Bado, L.; Garcez, L.M.; Quinnell, R.J. Heterogeneities in Leishmania infantum Infection: Using Skin Parasite Burdens to Identify Highly Infectious Dogs. PLoS Negl. Trop. Dis. 2014, 8, e2583. [Google Scholar] [CrossRef]
  29. Doehl, J.S.P.; Bright, Z.; Dey, S.; Davies, H.; Magson, J.; Brown, N.; Romano, A.; Dalton, J.E.; Pinto, A.I.; Pitchford, J.W.; et al. Skin parasite landscape determines host infectiousness in visceral leishmaniasis. Nat. Commun. 2017, 8, 57. [Google Scholar] [CrossRef] [Green Version]
  30. Borja, L.S.; de Sousa, O.M.F.; Solcà, M.D.S.; Bastos, L.A.; Bordoni, M.; Magalhães, J.T.; Larangeira, D.F.; Barrouin-Melo, S.M.; Fraga, D.B.M.; Veras, P.S.T. Parasite load in the blood and skin of dogs naturally infected by Leishmania infantum is correlated with their capacity to infect sand fly vectors. Vet. Parasitol. 2016, 229, 110–117. [Google Scholar] [CrossRef] [Green Version]
  31. Laurenti, M.D.; Rossi, C.N.; da Matta, V.L.R.; Tomokane, T.; Corbett, C.E.P.; Secundino, N.F.C.; Pimenta, P.F.P.; Marcondes, M. Asymptomatic dogs are highly competent to transmit Leishmania (Leishmania) infantum chagasi to the natural vector. Vet. Parasitol. 2013, 196, 296–300. [Google Scholar] [CrossRef]
  32. Molina, R.; Amela, C.; Nieto, J.; Andres, M.S.; González, F.; Castillo, J.; Lucientes, J.; Alvar, J. Infectivity of dogs naturally infected with Leishmania infantum to colonized Phlebotomus perniciosus. Trans. R. Soc. Trop. Med. Hyg. 1994, 88, 491–493. [Google Scholar] [CrossRef]
  33. Courtenay, O.; Quinnell, R.J.; Garcez, L.M.; Shaw, J.J.; Dye, C. Infectiousness in a Cohort of Brazilian Dogs: Why Culling Fails to Control Visceral Leishmaniasis in Areas of High Transmission. J. Infect. Dis. 2002, 186, 1314–1320. [Google Scholar] [CrossRef]
  34. da Costa-Val, A.P.; Cavalcanti, R.R.; Gontijo, N.D.F.; Michalick, M.S.M.; Alexander, B.; Williams, P.; Melo, M.N. Canine visceral leishmaniasis: Relationships between clinical status, humoral immune response, haematology and Lutzomyia (Lutzomyia) longipalpis infectivity. Vet. J. 2007, 174, 636–643. [Google Scholar] [CrossRef]
  35. Michalsky, M.; Rocha, M.F.; Lima, A.C.V.M.D.R.; França-Silva, J.C.; Pires, M.Q.; Oliveira, F.S.; Pacheco, R.S.; dos Santos, S.L.; Barata, R.A.; Romanha, J.; et al. Infectivity of seropositive dogs, showing different clinical forms of leishmaniasis, to Lutzomyia longipalpis phlebotomine sand flies. Vet. Parasitol. 2007, 147, 67–76. [Google Scholar] [CrossRef] [PubMed]
  36. Verçosa, B.; Lemos, C.; Mendonça, I.; Silva, S.; de Carvalho, S.; Goto, H.; Costa, F. Transmission potential, skin inflammatory response, and parasitism of symptomatic and asymptomatic dogs with visceral leishmaniasis. BMC Vet. Res. 2008, 4, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Soares, M.R.A.; de Mendonça, I.L.; Bonfim, J.M.D.; Rodrigues, J.A.; Werneck, G.L.; Costa, C.H.N. Canine visceral leishmaniasis in Teresina, Brazil: Relationship between clinical features and infectivity for sand flies. Acta Trop. 2011, 117, 6–9. [Google Scholar] [CrossRef] [PubMed]
  38. Magalhães-Junior, J.T.; Mota, T.F.; Porfirio-Passos, G.; Larangeira, D.F.; Franke, C.R.; Barrouin-Melo, S.M. Xenodiagnosis on dogs with visceral leishmaniasis: Canine and sand fly aspects related to the parasite transmission. Vet. Parasitol. 2016, 223, 120–126. [Google Scholar] [CrossRef] [PubMed]
  39. Travi, B.L.; Ferro, C.; Cadena, H.; Tabares, C.J.; Osorio, Y. Canine visceral leishmaniasis in Colombia: Relationship between clinical and parasitologic status and infectivity for sand flies. Am. J. Trop. Med. Hyg. 2001, 64, 119–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Quinnell, R.J.; Courtenay, O. Transmission, reservoir hosts and control of zoonotic visceral leishmaniasis. Parasitology 2009, 136, 1915–1934. [Google Scholar] [CrossRef]
  41. Valverde, J.; Paun, A.; Inbar, E.; Romano, A.; Lewis, M.; Ghosh, K.; Sacks, D. Increased Transmissibility of Leishmania donovani From the Mammalian Host to Vector Sand Flies After Multiple Exposures to Sand Fly Bites. J. Infect. Dis. 2017, 215, 1285–1293. [Google Scholar] [CrossRef] [Green Version]
  42. Vojtkova, B.; Frynta, D.; Spitzova, T.; Lestinova, T.; Votypka, J.; Volf, P.; Sadlova, J. Repeated Sand Fly Bites of Infected BALB/c Mice Enhance the Development of Leishmania Lesions. Front. Trop. Dis. 2021, 2, 745104. [Google Scholar] [CrossRef]
  43. Singh, O.P.; Tiwary, P.; Kushwaha, A.K.; Singh, S.K.; Singh, D.K.; Lawyer, P.; Rowton, E.; Chaubey, R.; Singh, A.K.; Rai, T.K.; et al. Xenodiagnosis to evaluate the infectiousness of humans to sandflies in an area endemic for visceral leishmaniasis in Bihar, India: A transmission-dynamics study. Lancet Microbe 2021, 2, e23–e31. [Google Scholar] [CrossRef]
  44. Kirstein, O.D.; Abbasi, I.; Horwitz, B.Z.; Skrip, L.; Hailu, A.; Jaffe, C.; Lin, L.; Prow, T.; Warburg, A. Minimally invasive microbiopsies: A novel sampling method for identifying asymptomatic, potentially infectious carriers of Leishmania donovani. Int. J. Parasitol. 2017, 47, 609–616. [Google Scholar] [CrossRef] [PubMed]
  45. Molina, R.; Cafiavate, C.; Cercenado, E.; Lagunat, F.; López-Vélez, R.; Alvar, J. Indirect xenodiagnosis of visceral leishmaniasis in 10 HIV-infected patients using colonized Phlebotomus perniciosus. AIDS 1994, 8, 277–278. [Google Scholar] [CrossRef] [PubMed]
  46. Molina, R.; Pulido, F.; Laguna, F.; Alvar, J.; López-Vélez, R.; Lohse, J.M. Infection of sand flies by humans coinfected with Leishmania infantum and human immunodeficiency virus. Am. J. Trop. Med. Hyg. 1999, 60, 51–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Molina, R.; Jiménez, M.; García-Martínez, J.; Martín, J.V.S.; Carrillo, E.; Sánchez, C.; Moreno, J.; Alves, F.; Alvar, J. Role of asymptomatic and symptomatic humans as reservoirs of visceral leishmaniasis in a Mediterranean context. PLoS Negl. Trop. Dis. 2020, 14, e0008253. [Google Scholar] [CrossRef]
  48. Zijlstra, E.E.; Alves, F.; Rijal, S.; Arana, B.; Alvar, J. Post-kala-azar dermal leishmaniasis in the Indian subcontinent: A threat to the South-East Asia Region Kala-azar Elimination Programme. PLoS Negl. Trop. Dis. 2017, 11, e0005877. [Google Scholar] [CrossRef] [Green Version]
  49. Molina, R.; Ghosh, D.; Carrillo, E.; Monnerat, S.; Bern, C.; Mondal, D.; Alvar, J. Infectivity of Post-Kala-azar Dermal Leishmaniasis Patients to Sand Flies: Revisiting a Proof of Concept in the Context of the Kala-azar Elimination Program in the Indian Subcontinent. Clin. Infect. Dis. 2017, 65, 150–153. [Google Scholar] [CrossRef]
  50. Das, N.K.; Kumar, P.; Chatterjee, M. Post kala-azar dermal leishmaniasis: Clinical features and differential diagnosis. Indian J. Dermatol. 2021, 66, 24–33. [Google Scholar] [CrossRef]
  51. Mondal, D.; Bern, C.; Ghosh, D.; Rashid, M.; Molina, R.; Chowdhury, R.; Nath, R.; Ghosh, P.; Chapman, L.; Alim, A.; et al. Quantifying the Infectiousness of Post-Kala-Azar Dermal Leishmaniasis Toward Sand Flies. Clin. Infect. Dis. 2018, 69, 251–258. [Google Scholar] [CrossRef] [Green Version]
  52. Kamhawi, S.; Serafim, T.D. Patchy Parasitized Skin Governs Leishmania donovani Transmission to Sand Flies. Trends Parasitol. 2017, 33, 748–750. [Google Scholar] [CrossRef]
  53. Elnaiem, D.A.; Ward, R.D.; Young, P.E. Development ofLeishmania chagasi (Kinetoplastida: Trypanosomatidae) in the second blood-meal of its vectorLutzomyia longipalpis (Diptera: Psychodidae). Parasitol. Res. 1994, 80, 414–419. [Google Scholar] [CrossRef]
  54. Doehl, J.S.P.; Ashwin, H.; Brown, N.; Romano, A.; Carmichael, S.; Pitchford, J.W.; Kaye, P.M. Spatial Point Pattern Analysis Identifies Mechanisms Shaping the Skin Parasite Landscape in Leishmania donovani Infection. Front. Immunol. 2021, 12, 795554. [Google Scholar] [CrossRef] [PubMed]
  55. Rodero, M.P.; Licata, F.; Poupel, L.; Hamon, P.; Khosrotehrani, K.; Combadiere, C.; Boissonnas, A. In Vivo Imaging Reveals a Pioneer Wave of Monocyte Recruitment into Mouse Skin Wounds. PLoS ONE 2014, 9, e108212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Trindade, M.A.B.; Silva, L.L.D.C.; Braz, L.M.A.; Amato, V.S.; Naafs, B.; Sotto, M.N. Post-kala-azar dermal leishmaniasis and leprosy: Case report and literature review. BMC Infect. Dis. 2015, 15, 543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Aslan, H.; Oliveira, F.; Meneses, C.; Castrovinci, P.; Gomes, R.; Teixeira, C.; Derenge, C.A.; Orandle, M.; Gradoni, L.; Oliva, G.; et al. New Insights into the Transmissibility of Leishmania infantum from Dogs to Sand Flies: Experimental Vector-Transmission Reveals Persistent Parasite Depots at Bite Sites. J. Infect. Dis. 2016, 213, 1752–1761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Gasim, S.; Elhassan, A.M.; Khalil, E.A.G.; Ismail, A.; Kadaru, A.M.Y.; Kharazmi, A.; Theander, T.G. High levels of plasma IL-10 and expression of IL-10 by keratinocytes during visceral leishmaniasis predict subsequent development of post-kala-azar dermal leishmaniasis. Clin. Exp. Immunol. 1998, 111, 64–69. [Google Scholar] [CrossRef]
  59. Dey, N.S.; Senaratne, S.; Somaratne, V.; Madarasinghe, N.P.; Seneviratne, B.; Forrester, S.; de Oca, M.M.; Reis, L.C.; Moulik, S.; Walrad, P.B.; et al. Early reduction in PD-L1 expression predicts faster treatment response in human cutaneous leishmaniasis. J. Clin. Investig. 2021, 131, e142765. [Google Scholar] [CrossRef]
  60. Esch, K.J.; Juelsgaard, R.; Martinez, P.A.; Jones, D.E.; Petersen, C.A. Programmed Death 1–Mediated T Cell Exhaustion during Visceral Leishmaniasis Impairs Phagocyte Function. J. Immunol. 2013, 191, 5542–5550. [Google Scholar] [CrossRef] [Green Version]
  61. Guarga, J.; Moreno, J.; Lucientes, J.; Gracia, M.; Peribáñez, M.; Alvar, J.; Castillo, J. Canine leishmaniasis transmission: Higher infectivity amongst naturally infected dogs to sand flies is associated with lower proportions of T helper cells. Res. Vet. Sci. 2000, 69, 249–253. [Google Scholar] [CrossRef]
  62. Schaut, R.G.; Lamb, I.M.; Toepp, A.J.; Scott, B.; Mendes-Aguiar, C.O.; Coutinho, J.F.V.; Jeronimo, S.M.B.; Wilson, M.; Harty, J.T.; Waldschmidt, T.J.; et al. Regulatory IgDhiB Cells Suppress T Cell Function via IL-10 and PD-L1 during Progressive Visceral Leishmaniasis. J. Immunol. 2016, 196, 4100–4109. [Google Scholar] [CrossRef] [Green Version]
  63. Cabral, S.M.; Silvestre, R.L.; Santarém, N.M.; Tavares, J.C.; Silva, A.F.; Cordeiro-Da-Silva, A. A Leishmania infantum cytosolic tryparedoxin activates B cells to secrete interleukin-10 and specific immunoglobulin. Immunology 2008, 123, 555–565. [Google Scholar] [CrossRef]
  64. Belkaid, Y.; Hoffmann, K.F.; Mendez, S.; Kamhawi, S.; Udey, M.C.; Wynn, T.; 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]
  65. Sandoval, C.; Araujo, G.; Sosa, W.; Avalos, S.; Silveira, F.; Corbett, C.; Zúniga, C.; Laurenti, M. In situ cellular immune response in non-ulcerated skin lesions due to Leishmania (L.) infantum chagasi infection. J. Venom. Anim. Toxins Incl. Trop. Dis. 2021, 27, e20200149. [Google Scholar] [CrossRef] [PubMed]
  66. Ordeix, L.; Montserrat-Sangrà, S.; Martínez-Orellana, P.; Solano-Gallego, L. Toll-Like Receptors 2, 4, and 7, Interferon-Gamma, Interleukin 10, and Programmed Death Ligand 1 Transcripts in Leishmanin Skin Test-Positive Reactions of Ibizan Hound Dogs. J. Immunol. Res. 2020, 2020, 9602576. [Google Scholar] [CrossRef] [PubMed]
  67. Hernandez-Bures, A.; Gillen, J.; Apostolidis, K.; Saridomichelakis, M.; Di Loria, A.; Santoro, D. Evaluation of the cutaneous inflammatory cells in dogs with leishmaniosis and in dogs without the disease that were naturally infected by Leishmania infantum (syn. L. chagasi). Vet. Dermatol. 2020, 32, 99. [Google Scholar] [CrossRef]
  68. Rodrigues, O.R.; Marques, C.; Soares-Clemente, M.; Ferronha, M.H.; Santos-Gomes, G.M. Identification of regulatory T cells during experimental Leishmania infantum infection. Immunobiology 2009, 214, 101–111. [Google Scholar] [CrossRef]
  69. Borbón, T.Y.; Scorza, B.M.; Clay, G.M.; De Queiroz, F.L.N.; Sariol, A.J.; Bowen, J.L.; Chen, Y.; Zhanbolat, B.; Parlet, C.P.; Valadares, D.G.; et al. Coinfection with Leishmania major and Staphylococcus aureus enhances the pathologic responses to both microbes through a pathway involving IL-17A. PLoS Negl. Trop. Dis. 2019, 13, e0007247. [Google Scholar] [CrossRef]
  70. 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] [Green Version]
  71. Guimaraes-Costa, A.B.; Shannon, J.P.; Waclawiak, I.; Oliveira, J.; Meneses, C.; de Castro, W.; Wen, X.; Brzostowski, J.; Serafim, T.D.; Andersen, J.F.; et al. A sand fly salivary protein acts as a neutrophil chemoattractant. Nat. Commun. 2021, 12, 3213. [Google Scholar] [CrossRef]
  72. Dey, R.; Joshi, A.B.; Oliveira, F.; Pereira, L.; Guimarães-Costa, A.B.; Serafim, T.D.; de Castro, W.; Coutinho-Abreu, I.; 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 2017, 23, 134–143.e6. [Google Scholar] [CrossRef] [Green Version]
  73. Thalhofer, C.J.; Chen, Y.; Sudan, B.; Love-Homan, L.; Wilson, M.E. Leukocytes Infiltrate the Skin and Draining Lymph Nodes in Response to the Protozoan Leishmania infantum chagasi. Infect. Immun. 2011, 79, 108–117. [Google Scholar] [CrossRef] [Green Version]
  74. Atayde, V.D.; Hassani, K.; Filho, A.D.S.L.; Borges, A.R.; Adhikari, A.; Martel, C.; Olivier, M. Leishmania exosomes and other virulence factors: Impact on innate immune response and macrophage functions. Cell. Immunol. 2016, 309, 7–18. [Google Scholar] [CrossRef] [PubMed]
  75. Rogers, M.; Kropf, P.; Choi, B.-S.; Dillon, R.; Podinovskaia, M.; Bates, P.; Müller, I. Proteophosophoglycans Regurgitated by Leishmania-Infected Sand Flies Target the L-Arginine Metabolism of Host Macrophages to Promote Parasite Survival. PLoS Pathog. 2009, 5, e1000555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. van der Zande, H.J.P.; Nitsche, D.; Schlautmann, L.; Guigas, B.; Burgdorf, S. The Mannose Receptor: From Endocytic Receptor and Biomarker to Regulator of (Meta)Inflammation. Front. Immunol. 2021, 12, 765034. [Google Scholar] [CrossRef] [PubMed]
  77. Lee, S.H.; Charmoy, M.; Romano, A.; Paun, A.; Chaves, M.M.; Cope, F.O.; Ralph, D.A.; Sacks, D.L. Mannose receptor high, M2 dermal macrophages mediate nonhealing Leishmania major infection in a Th1 immune environment. J. Exp. Med. 2018, 215, 357–375. [Google Scholar] [CrossRef] [PubMed]
  78. Jenkins, S.J.; Ruckerl, D.; Thomas, G.; Hewitson, J.; Duncan, S.; Brombacher, F.; Maizels, R.M.; Hume, D.; Allen, J. IL-4 directly signals tissue-resident macrophages to proliferate beyond homeostatic levels controlled by CSF-1. J. Exp. Med. 2013, 210, 2477–2491. [Google Scholar] [CrossRef] [PubMed]
  79. Chaves, M.M.; Lee, S.H.; Kamenyeva, O.; Ghosh, K.; Peters, N.C.; Sacks, D. The role of dermis resident macrophages and their interaction with neutrophils in the early establishment of Leishmania major infection transmitted by sand fly bite. PLoS Pathog. 2020, 16, e1008674. [Google Scholar] [CrossRef]
  80. Pacheco, C.M.S.; Flores, G.V.A.; Gonzalez, K.; Gomes, C.M.D.C.; Passero, L.F.D.; Tomokane, T.Y.; Sosa-Ochoa, W.; Zúniga, C.; Calzada, J.; Saldaña, A.; et al. Macrophage Polarization in the Skin Lesion Caused by Neotropical Species of Leishmania sp. J. Immunol. Res. 2021, 2021, 5596876. [Google Scholar] [CrossRef]
  81. Veloso, L.B.; Cardoso, F.D.O.; Charret, K.D.S.; Silva, M.P.D.S.; Côrtes, L.M.D.C.; Calabrese, K.D.S.; da Silva, F.S.; de Sousa, J.F.; Rocha, M.F.; Alves, C.R. Detection of Metalloproteases and Cysteine Proteases RNA Transcripts of Leishmania (Leishmania) infantum in Ear Edge Skin of Naturally Infected Dogs. BioMed Res. Int. 2020, 2020, 2615787. [Google Scholar] [CrossRef]
  82. DeSouza-Vieira, T.; Iniguez, E.; Serafim, T.D.; De Castro, W.; Karmakar, S.; Disotuar, M.M.; Cecilio, P.; Lacsina, J.R.; Meneses, C.; Nagata, B.M.; et al. Heme Oxygenase-1 Induction by Blood-Feeding Arthropods Controls Skin Inflammation and Promotes Disease Tolerance. Cell Rep. 2020, 33, 108317. [Google Scholar] [CrossRef]
  83. Luz, N.F.; DeSouza-Vieira, T.; De Castro, W.; Vivarini, A.C.; Pereira, L.; França, R.R.; Silveira-Mattos, P.S.; Costa, D.; Teixeira, C.; Meneses, C.; et al. Lutzomyia longipalpis Saliva Induces Heme Oxygenase-1 Expression at Bite Sites. Front. Immunol. 2018, 9, 2779. [Google Scholar] [CrossRef]
  84. Collin, N.; Gomes, R.; Teixeira, C.; Cheng, L.; Laughinghouse, A.; Ward, J.M.; Elnaiem, D.-E.; Fischer, L.; Valenzuela, J.G.; Kamhawi, S. Sand Fly Salivary Proteins Induce Strong Cellular Immunity in a Natural Reservoir of Visceral Leishmaniasis with Adverse Consequences for Leishmania. PLoS Pathog. 2009, 5, e1000441. [Google Scholar] [CrossRef] [PubMed]
  85. Ribeiro, J.M.; Modi, G. The Salivary Adenosine/AMP Content of Phlebotomus Argentipes Annandale and Brunetti, the Main Vector of Human Kala-Azar. J. Parasitol. 2001, 87, 915–917. [Google Scholar] [CrossRef]
  86. Lerner, E.; Ribeiro, J.; Nelson, R.; Lerner, M. Isolation of maxadilan, a potent vasodilatory peptide from the salivary glands of the sand fly Lutzomyia longipalpis. J. Biol. Chem. 1991, 266, 11234–11236. [Google Scholar] [CrossRef]
  87. Morris, R.V.; Shoemaker, C.B.; David, J.R.; Lanzaro, G.C.; Titus, R.G. Sandfly Maxadilan Exacerbates Infection with Leishmania major and Vaccinating Against It Protects Against L. major Infection. J. Immunol. 2001, 167, 5226–5230. [Google Scholar] [CrossRef] [Green Version]
  88. Abdeladhim, M.; Kamhawi, S.; Valenzuela, J.G. What’s behind a sand fly bite? The profound effect of sand fly saliva on host hemostasis, inflammation and immunity. Infect. Genet. Evol. 2014, 28, 691–703. [Google Scholar] [CrossRef] [Green Version]
  89. 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]
  90. Scorza, B.M.; Wacker, M.A.; Messingham, K.; Kim, P.; Klingelhutz, A.; Fairley, J.; Wilson, M.E. Differential Activation of Human Keratinocytes by Leishmania Species Causing Localized or Disseminated Disease. J. Investig. Dermatol. 2017, 137, 2149–2156. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Outward parasite transmission to naïve sand flies in a model of chronic L. infantum infection. Parasitized macrophages cluster together in the skin in a self-propagating manner and heterogenous, patchy dermal distribution. Establishment of chronic infection in the skin is promoted by decreased CD4+ T cell effector responses, decreased IFNy secretion, increased PD-L1, hypergammaglobulinemia, and increased B-cell IL-10 secretion. When naïve sand flies feed, amastigote-parasitized macrophages are transmitted from the skin into the sand fly gut, infecting the sand fly. The bite site receives sand fly salivary proteins, sand fly gut microbiota, and an increase in erythrocyte heme-oxygenase-1 production, all of which contribute to an anti-inflammatory environment and subsequent parasite persistence. Created with BioRender.com, accessed on 10 March 2022.
Figure 1. Outward parasite transmission to naïve sand flies in a model of chronic L. infantum infection. Parasitized macrophages cluster together in the skin in a self-propagating manner and heterogenous, patchy dermal distribution. Establishment of chronic infection in the skin is promoted by decreased CD4+ T cell effector responses, decreased IFNy secretion, increased PD-L1, hypergammaglobulinemia, and increased B-cell IL-10 secretion. When naïve sand flies feed, amastigote-parasitized macrophages are transmitted from the skin into the sand fly gut, infecting the sand fly. The bite site receives sand fly salivary proteins, sand fly gut microbiota, and an increase in erythrocyte heme-oxygenase-1 production, all of which contribute to an anti-inflammatory environment and subsequent parasite persistence. Created with BioRender.com, accessed on 10 March 2022.
Pathogens 11 00610 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

Arumugam, S.; Scorza, B.M.; Petersen, C. Visceral Leishmaniasis and the Skin: Dermal Parasite Transmission to Sand Flies. Pathogens 2022, 11, 610. https://0-doi-org.brum.beds.ac.uk/10.3390/pathogens11060610

AMA Style

Arumugam S, Scorza BM, Petersen C. Visceral Leishmaniasis and the Skin: Dermal Parasite Transmission to Sand Flies. Pathogens. 2022; 11(6):610. https://0-doi-org.brum.beds.ac.uk/10.3390/pathogens11060610

Chicago/Turabian Style

Arumugam, Sahaana, Breanna M. Scorza, and Christine Petersen. 2022. "Visceral Leishmaniasis and the Skin: Dermal Parasite Transmission to Sand Flies" Pathogens 11, no. 6: 610. https://0-doi-org.brum.beds.ac.uk/10.3390/pathogens11060610

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop