Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.557
Filter
1.
J Vis Exp ; (211)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39345111

ABSTRACT

Immuno-metabolism is a pivotal determinant in the progression of leishmaniasis. Synthetic biology-based approach has garnered significant attention as a step toward therapeutic intervention targeting host-associated factors that drive leishmaniasis. Synthetic biology entails the engineering of genetic components in an orthogonal and modular manner to precisely modulate biological systems, imparting novel functions to cells. In the presented study, elucidation of a systematic pipeline for the development of an inducible tetracycline-controlled (TetON)-based synthetic circuit was aimed at delivering succinate dehydrogenase as a therapeutic agent to facilitate the elimination of intracellular Leishmania parasites. The outlined protocol describes the designing of a synthetic circuit and its subsequent validation. The proposed strategy also concentrates on the incorporation of synthetic circuits in the plasmid backbone as a delivery vehicle. Additionally, delivery machinery employing polyplexes-based nano-particles for the delivery of synthetic circuits was used in murine macrophage cell lines without compromising the cellular morphology. Standardization of the method was conducted for selecting transfected cells and determining optimal induction concentration for synthetic circuit expression. Observations reveal a distinct reduction in intracellular parasite burden in transfected cells compared to infected cells. Pro-inflammatory cytokines were expressed post-infection in synthetic circuit transfected and induced cells as a mechanism to promote parasite elimination. This underscores the synthetic biology-based method as a potent approach in leishmaniasis by targeting host factors associated with disease progression.


Subject(s)
Leishmaniasis , Mice , Animals , Leishmaniasis/immunology , Leishmaniasis/therapy , Leishmaniasis/parasitology , Synthetic Biology/methods , Leishmania/immunology , Macrophages/parasitology , Macrophages/metabolism , Macrophages/immunology , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism , Plasmids/genetics
2.
Biomolecules ; 14(8)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39199338

ABSTRACT

Type 2 diabetes mellitus is a metabolic disorder that causes chronic high blood sugar levels, and diabetic patients are more susceptible to infections. American cutaneous leishmaniasis is an infectious disease caused by a parasite that affects the skin and mucous membranes, leading to one or multiple ulcerative lesions. Chronic inflammation and functional changes in various organs and systems, including the immune system, are the primary causes of both diseases. Melatonin, an essential immunomodulatory, antioxidant, and neuroprotective agent, can benefit many immunological processes and infectious diseases, including leishmaniasis. Although, limited reports are available on diabetic patients with leishmaniasis. The literature suggests that melatonin may play a promising role in inflammatory disorders. This study was designed to assess melatonin levels and inflammatory mediators in diabetic patients affected by leishmaniasis. Blood samples from 25 individuals were analyzed and divided into four groups: a control group (without any diseases), a Leishmania-positive group, patients with type 2 diabetes mellitus, and patients with a combination of both diseases. This study measured the serum levels of melatonin through ELISA, while IL-4 and TNF-α were measured using flow cytometry, and C-reactive protein was measured through turbidimetry. This study found that patients with leishmaniasis significantly increased TNF-α and decreased melatonin levels. However, the group of diabetic patients with leishmaniasis showed higher melatonin levels than the control group. These observations suggest that TNF-α may influence melatonin production in patients with American cutaneous leishmaniasis, potentially contributing to the inflammatory characteristics of both diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Inflammation , Melatonin , Tumor Necrosis Factor-alpha , Melatonin/blood , Melatonin/metabolism , Humans , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/immunology , Male , Female , Middle Aged , Hyperglycemia/metabolism , Hyperglycemia/blood , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism , Inflammation/metabolism , Inflammation/blood , Adult , Interleukin-4/blood , Leishmaniasis, Cutaneous/blood , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/metabolism , C-Reactive Protein/metabolism , Leishmaniasis/blood , Leishmaniasis/immunology , Leishmaniasis/metabolism , Leishmaniasis/parasitology , Aged
3.
Front Biosci (Landmark Ed) ; 29(8): 292, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39206906

ABSTRACT

As professional phagocytes, macrophages represent the first line of defence against invading microbial pathogens. Various cellular processes such as programmed cell death, autophagy and RNA interference (RNAi) of macrophages are involved directly in elimination or assist in elimination of invading pathogens. However, parasites, such as Leishmania, have evolved diverse strategies to interfere with macrophage cell functions, favouring their survival, growth and replication inside hostile and restrictive environments of macrophages. Therefore, identification and detailed characterization of macrophage-pathogen interactions is the key to understanding how pathogens subvert macrophage functions to support their infection and disease process. In recent years, great progress has been achieved in understanding how Leishmania affects with critical host macrophage functions. Based on latest progress and accumulating knowledge, this review exclusively focuses on macrophage-Leishmania interaction, providing an overview of macrophage cellular processes such as programmed cell death, autophagy and RNAi during Leishmania infection. Despite extensive progress, many questions remain and require further investigation.


Subject(s)
Autophagy , Leishmania , Leishmaniasis , Macrophages , Leishmaniasis/immunology , Leishmaniasis/parasitology , Humans , Macrophages/immunology , Macrophages/parasitology , Leishmania/immunology , Animals , RNA Interference , Apoptosis
4.
Front Immunol ; 15: 1437430, 2024.
Article in English | MEDLINE | ID: mdl-39211053

ABSTRACT

Macrophages play a pivotal role as host cells for Leishmania parasites, displaying a notable functional adaptability ranging from the proinflammatory, leishmanicidal M1 phenotype to the anti-inflammatory, parasite-permissive M2 phenotype. While macrophages can potentially eradicate amastigotes through appropriate activation, Leishmania employs diverse strategies to thwart this activation and redirect macrophages toward an M2 phenotype, facilitating its survival and replication. Additionally, a competition for iron between the two entities exits, as iron is vital for both and is also implicated in macrophage defensive oxidative mechanisms and modulation of their phenotype. This review explores the intricate interplay between macrophages, Leishmania, and iron. We focus the attention on the potential of iron oxide nanoparticles (IONPs) as a sort of immunotherapy to treat some leishmaniasis forms by reprogramming Leishmania-permissive M2 macrophages into antimicrobial M1 macrophages. Through the specific targeting of iron in macrophages, the use of IONPs emerges as a promising strategy to finely tune the parasite-host interaction, endowing macrophages with an augmented antimicrobial arsenal capable of efficiently eliminating these intrusive microbes.


Subject(s)
Leishmania , Leishmaniasis , Macrophage Activation , Macrophages , Magnetic Iron Oxide Nanoparticles , Macrophages/immunology , Macrophages/metabolism , Humans , Leishmaniasis/immunology , Leishmaniasis/drug therapy , Animals , Macrophage Activation/drug effects , Macrophage Activation/immunology , Leishmania/immunology , Leishmania/drug effects , Host-Parasite Interactions/immunology
5.
Trends Parasitol ; 40(9): 788-804, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39174373

ABSTRACT

Leishmania is an intracellular protozoan transmitted by sand fly vectors; it causes cutaneous, mucocutaneous, or visceral disease. Its growth and survival are impeded by type 1 T helper cell responses, which entail interferon (IFN)-γ-mediated macrophage activation. Leishmania partially escapes this host defense by triggering immune cell and cytokine responses that favor parasite replication rather than killing. Novel methods for in situ analyses have revealed that the pathways of immune control and microbial evasion are strongly influenced by the tissue context, the micro milieu factors, and the metabolism at the site of infection, which we collectively term the 'immunomicrotope'. Understanding the components and the impact of the immunomicrotope will enable the development of novel strategies for the treatment of chronic leishmaniasis.


Subject(s)
Leishmania , Leishmaniasis , Leishmania/immunology , Animals , Humans , Leishmaniasis/immunology , Immune Evasion/immunology , Host-Parasite Interactions/immunology
6.
Immunol Lett ; 268: 106885, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38901739

ABSTRACT

Leishmaniasis is a collective term for several tropical, neglected diseases caused by protozoans of the species Leishmania, 20 of which causing disease in humans ranging from localised self-healing lesions to chronic manifestations which affect the skin or inner organs. Although millions of infections are accounted for annually, treatment options are scarce and limited to medication associated with heavy side-effects and increasing antibiotic resistance. Case studies point towards immunotherapy as effective alternative treatment relying on immunomodulatory properties of e.g., the Bacillus Calmette-Guérin vaccine. Leishmania parasites are also known to modulate the immune system, yet the underlying macromolecules and surface molecules remain widely under characterised. With this short review, we aim to provide a complete summary of the existing literature describing one of the most expressed surface molecule on Leishmania spp, lipophosphoglycan (LPG), which shows great variability between different lifecycle stages and different Leishmania spp. Complete characterisation of LPG may aid to improve treatment and aid the development of vaccination strategies, and open new avenues to exploit the immunomodulatory properties of LPG in unrelated conditions.


Subject(s)
Glycosphingolipids , Immunomodulation , Leishmania , Leishmaniasis , Leishmania/immunology , Humans , Glycosphingolipids/immunology , Glycosphingolipids/metabolism , Animals , Leishmaniasis/immunology , Leishmaniasis/parasitology
7.
Microbes Infect ; 26(5-6): 105353, 2024.
Article in English | MEDLINE | ID: mdl-38763478

ABSTRACT

The obligate intracellular parasite Leishmania binds several receptors to trigger uptake by phagocytic cells, ultimately resulting in visceral or cutaneous leishmaniasis. A series of signaling pathways in host cells, which are critical for establishment and persistence of infection, are activated during Leishmania internalization. Thus, preventing Leishmania uptake by phagocytes could be a novel therapeutic strategy for leishmaniasis. However, the host cellular machinery mediating promastigote and amastigote uptake is not well understood. Here, using small molecule inhibitors of Mitogen-activated protein/Extracellular signal regulated kinases (MAPK/ERK), we demonstrate that ERK1/2 mediates Leishmania amazonensis uptake and (to a lesser extent) phagocytosis of beads by macrophages. We find that inhibiting host MEK1/2 or ERK1/2 leads to inefficient amastigote uptake. Moreover, using inhibitors and primary macrophages lacking spleen tyrosine kinase (SYK) or Abl family kinases, we show that SYK and Abl family kinases mediate Raf, MEK, and ERK1/2 activity and are necessary for uptake. Finally, we demonstrate that trametinib, a MEK1/2 inhibitor used to treat cancer, reduces disease severity and parasite burden in Leishmania-infected mice, even if it is started after lesions develop. Our results show that maximal Leishmania infection requires MAPK/ERK and highlight potential for MAPK/ERK-mediated signaling pathways to be novel therapeutic targets for leishmaniasis.


Subject(s)
Macrophages , Animals , Macrophages/parasitology , Macrophages/metabolism , Macrophages/immunology , Mice , Phagocytosis , Pyridones/pharmacology , Leishmaniasis/parasitology , Leishmaniasis/immunology , Syk Kinase/metabolism , Syk Kinase/antagonists & inhibitors , MAP Kinase Signaling System , Mice, Inbred C57BL , Leishmania mexicana/enzymology , Leishmania , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/metabolism , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/pathology , Pyrimidinones
8.
Cell Immunol ; 399-400: 104826, 2024.
Article in English | MEDLINE | ID: mdl-38669897

ABSTRACT

Infectious diseases like leishmaniasis, malaria, HIV, tuberculosis, leprosy and filariasis are responsible for an immense burden on public health systems. Among these, leishmaniasis is under the category I diseases as it is selected by WHO (World Health Organization) on the ground of diversity and complexity. High cost, resistance and toxic effects of Leishmania traditional drugs entail identification and development of therapeutic alternative. Since the natural infection elicits robust immunity, consistence efforts are going on to develop a successful vaccine. Clinical trials have been conducted on vaccines like Leish-F1, F2, and F3 formulated using specific Leishmania antigen epitopes. Current strategies utilize individual or combined antigens from the parasite or its insect vector's salivary gland extract, with or without adjuvant formulation for enhanced efficacy. Promising animal data supports multiple vaccine candidates (Lmcen-/-, LmexCen-/-), with some already in or heading for clinical trials. The crucial challenge in Leishmania vaccine development is to translate the research knowledge into affordable and accessible control tools that refines the outcome for those who are susceptible to infection. This review focuses on recent findings in Leishmania vaccines and highlights difficulties facing vaccine development and implementation.


Subject(s)
Leishmania , Leishmaniasis Vaccines , Leishmaniasis , Vaccine Development , Humans , Leishmaniasis Vaccines/immunology , Animals , Leishmania/immunology , Leishmaniasis/immunology , Leishmaniasis/prevention & control , Vaccine Development/methods , Antigens, Protozoan/immunology , Clinical Trials as Topic
9.
Biol Trace Elem Res ; 202(1): 210-220, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37088826

ABSTRACT

In leishmaniasis, the protective immunity is largely mediated by proinflammatory cytokine producing abilities of T cells and an efficient parasite killing by phagocytic cells. Notwithstanding a substantial progress that has been made during last decades, the mechanisms or factors involved in establishing protective immunity against Leishmania are not identified. In ancient Indian literature, metallic "bhasma," particularly that of "swarna" or gold (fine gold particles), is indicated as one of the most prominent metal-based therapeutic medicine, which is known to impart protective and curative properties in various health issues. In this work, we elucidated the potential of swarna bhasma (SB) on the effector properties of phagocytes and antigen-activated CD4+ T cells in augmenting the immunogenicity of L. donovani antigens. The characterization of SB revealing its shape, size, composition, and measurement of cytotoxicity established the physiochemical potential for its utilization as an immunomodulator. The activation of macrophages with SB enhanced their capacity to produce nitric oxide and proinflammatory cytokines, which eventually resulted in reduced uptake of parasites and their proliferation in infected cells. Further, in Leishmania-infected animals, SB administration reduced the generation of IL-10, an anti-inflammatory cytokine, and enhanced pro-inflammatory cytokine generation by antigen activated CD4+ T cells with increased frequency of double (IFNγ+/TNFα+) and triple (IFNγ+TNFα+IL-2+) positive cells and abrogated disease pathogeneses at the early days of infection. Our results also suggested that cow-ghee (A2) emulsified preparation of SB, either alone or with yashtimadhu, a known natural immune modulator which enhances the SB's potential in enhancing the immunogenicity of parasitic antigens. These findings suggested a definite potential of SB in enhancing the effector functions of phagocytes and CD4+ T cells against L. donovani antigens. Therefore, more studies are needed to elucidate the mechanistic details of SB and its potential in enhancing vaccine-induced immunity.


Subject(s)
Antigen Presentation , Antigens, Protozoan , CD4-Positive T-Lymphocytes , Calotropis , Gold , Latex , Leishmania donovani , Macrophages , Medicine, Ayurvedic , Th1 Cells , Arsenic , Drug Combinations , Gold/administration & dosage , Gold/pharmacology , Latex/administration & dosage , Latex/pharmacology , Lead , Macrophages/drug effects , Macrophages/immunology , CD4-Positive T-Lymphocytes/immunology , Phagocytes/drug effects , Phagocytes/immunology , Leishmaniasis/immunology , Leishmaniasis/parasitology , Leishmania donovani/drug effects , Leishmania donovani/growth & development , Leishmania donovani/immunology , Antigens, Protozoan/immunology , Th1 Cells/immunology , Animals , Mice , RAW 264.7 Cells , Female , Mice, Inbred BALB C
10.
Front Cell Infect Microbiol ; 12: 941888, 2022.
Article in English | MEDLINE | ID: mdl-35992159

ABSTRACT

Leishmania RNA virus 1 (LRV1) is a double-stranded RNA virus found in some strains of the human protozoan parasite Leishmania, the causative agent of leishmaniasis, a neglected tropical disease. Interestingly, the presence of LRV1 inside Leishmania constitutes an important virulence factor that worsens the leishmaniasis outcome in a type I interferon (IFN)-dependent manner and contributes to treatment failure. Understanding how macrophages respond toward Leishmania alone or in combination with LRV1 as well as the role that type I IFNs may play during infection is fundamental to oversee new therapeutic strategies. To dissect the macrophage response toward infection, RNA sequencing was performed on murine wild-type and Ifnar-deficient bone marrow-derived macrophages infected with Leishmania guyanensis (Lgy) devoid or not of LRV1. Additionally, macrophages were treated with poly I:C (mimetic virus) or with type I IFNs. By implementing a weighted gene correlation network analysis, the groups of genes (modules) with similar expression patterns, for example, functionally related, coregulated, or the members of the same functional pathway, were identified. These modules followed patterns dependent on Leishmania, LRV1, or Leishmania exacerbated by the presence of LRV1. Not only the visualization of how individual genes were embedded to form modules but also how different modules were related to each other were observed. Thus, in the context of the observed hyperinflammatory phenotype associated to the presence of LRV1, it was noted that the biomarkers tumor-necrosis factor α (TNF-α) and the interleukin 6 (IL-6) belonged to different modules and that their regulating specific Src-family kinases were segregated oppositely. In addition, this network approach revealed the strong and sustained effect of LRV1 on the macrophage response and genes that had an early, late, or sustained impact during infection, uncovering the dynamics of the IFN response. Overall, this study contributed to shed light and dissect the intricate macrophage response toward infection by the Leishmania-LRV1 duo and revealed the crosstalk between modules made of coregulated genes and provided a new resource that can be further explored to study the impact of Leishmania on the macrophage response.


Subject(s)
Interferon Type I , Leishmania , Leishmaniasis , Leishmaniavirus , Macrophages , Animals , Humans , Interferon Type I/immunology , Leishmania/virology , Leishmaniasis/immunology , Leishmaniasis/parasitology , Leishmaniasis/virology , Macrophages/immunology , Macrophages/parasitology , Mice
11.
Front Immunol ; 13: 762080, 2022.
Article in English | MEDLINE | ID: mdl-35145518

ABSTRACT

Leishmania spp. infection outcomes are dependent on both host and parasite factors. Manipulation of host signaling pathways involved in the generation of immune responses is thought to be one of the most common mechanisms used by parasites for persistence within the host. Considering the diversity of pathologies caused by different Leishmania spp., it is plausible that significant differences may exist in the mechanisms of host cell manipulation by each parasite species, which may have implications when developing new vaccine or treatment strategies. Here we show that in L. braziliensis-infection in BALB/c mice, a model of resistance, activation of ERK1/2 coincides with the peak of inflammatory responses and resolution of tissue parasitism. In contrast, in the susceptibility model of L. amazonensis-infection, an early silent phase of infection is observed, detected solely by quantification of parasite loads. At this early stage, only basal levels of P-ERK1/2 are observed. Later, after a brief shutdown of ERK1/2 phosphorylation, disease progression is observed and is associated with increased inflammation, lesion size and tissue parasitism. Moreover, the short-term down-regulation of ERK1/2 activation affected significantly downstream inflammatory pathways and adaptive T cell responses. Administration of U0126, a MEK/ERK inhibitor, confirmed this phenomenon, since bigger lesions and higher parasite loads were seen in infected mice that received U0126. To investigate how kinetics of ERK1/2 activation could affect the disease progression, U0126 was administered to L. amazonensis-infected animals earlier than the P-ERK1/2 switch off time-point. This intervention resulted in anticipation of the same effects on inflammatory responses and susceptibility phenotype seen in the natural course of infection. Additionally, in vitro inhibition of ERK1/2 affected the phagocytosis of L. amazonensis by BMDMs. Collectively, our findings reveal distinct temporal patterns of activation of inflammatory responses in L. braziliensis and L. amazonensis in the same animal background and a pivotal role for a brief and specific shutdown of ERK1/2 activation at late stages of L. amazonensis infection. Since activation of inflammatory responses is a crucial aspect for the control of infectious processes, these findings may be important for the search of new and specific strategies of vaccines and treatment for tegumentary leishmaniasis.


Subject(s)
Immunity, Cellular , Leishmania mexicana/immunology , Leishmaniasis/immunology , Leishmaniasis/metabolism , Leishmaniasis/parasitology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Host-Pathogen Interactions/immunology , Inflammation Mediators/metabolism , Leishmaniasis/pathology , Mice , Parasite Load , Phagocytosis/immunology , Phosphorylation , Signal Transduction
12.
Infect Genet Evol ; 98: 105210, 2022 03.
Article in English | MEDLINE | ID: mdl-35031509

ABSTRACT

γδ T cells are thymus derived heterogeneous and unconventional T- lymphocyte expressing TCR γ (V γ9) and TCRδ (Vδ2) chain and play an important role in connecting innate and adaptive armaments of immune response. These cells can recognize wide ranges of antigens even without involvement of major histocompatibility complex and exert their biological functions by cytotoxicity or activating various types of immune cells. In recent past, γδ T cells have emerged as an important player during protozoa infection and rapidly expand after exposure with them. They have also been widely studied in vaccine induced immune response against many bacterial and protozoan infections with improved clinical outcome. In this review, we will discuss the various roles of γδ T cells in immunity against malaria and leishmaniasis, the two important protozoan diseases causing significant mortality and morbidity throughout the world.


Subject(s)
Immunity, Innate , Intraepithelial Lymphocytes/immunology , Leishmaniasis/immunology , Malaria/immunology , Humans
13.
Biomed J ; 45(1): 109-117, 2022 02.
Article in English | MEDLINE | ID: mdl-34175493

ABSTRACT

Leishmaniasis is a neglected tropical disease that causes several clinical manifestations. Parasites of the genus Leishmania cause this disease. Spread across five continents, leishmaniasis is a particular public health problem in developing countries. Leishmania infects phagocytic cells such as macrophages, where it induces adenosine triphosphate (ATP) release at the time of infection. ATP activates purinergic receptors in the cell membranes of infected cells and promotes parasite control by inducing leukotriene B4 release and NLRP3 inflammasome activation. Moreover, uridine triphosphate induces ATP release, exacerbating the immune response. However, ATP may also undergo catalysis by ectonucleotidases present in the parasite membrane, generating adenosine, which activates P1 receptors and induces the production of anti-inflammatory molecules such as prostaglandin E2 and IL-10. These mechanisms culminate in Leishmania's survival. Thus, how Leishmania handles extracellular nucleotides and the activation of purinergic receptors determines the control or the dissemination of the disease.


Subject(s)
Leishmania , Leishmaniasis , Receptors, Purinergic , Adenosine , Adenosine Triphosphate , Dinoprostone/immunology , Humans , Interleukin-10/immunology , Leishmania/physiology , Leishmaniasis/immunology , Leukotriene B4/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Receptors, Purinergic/metabolism , Signal Transduction
14.
Int Immunopharmacol ; 102: 108400, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34890999

ABSTRACT

Leishmaniasis, a neglected tropical disease, still remains a global concern for the healthcare sector. The primary causative agents of the disease comprise diverse leishmanial species, leading to recurring failures in disease diagnosis and delaying the initiation of appropriate chemotherapy. Various species of the Leishmania parasite cause diverse clinical manifestations ranging from skin ulcers to systemic infections. Therefore, host immunity in response to different forms of infecting species of Leishmania becomes pivotal in disease progression or regression. Thus, understanding the paradox of immune arsenals during host and parasite interface becomes crucial to eliminate this deadly disease. In the present review, we have elaborated on the immunological perspectives of the disease and discussed primary host immune cells that form a defense line to counteract parasite infection. Furthermore, we also have shed light on the immune cells and effector molecules responsible for parasite survival in host lethal milieu/ environment. Next, we have highlighted recent molecules/compounds showing potent leishmanicidal activities pertaining to their pro-oxidant and immuno-modulatory mechanisms. This review addresses an immuno-biological overview of the factors influencing the parasitic disease, as this knowledge can aid in the unraveling/ identification of potential biomarkers, novel therapeutics, and vaccine candidates against leishmaniasis.


Subject(s)
Leishmania/immunology , Leishmaniasis/immunology , Animals , Host-Parasite Interactions/immunology , Humans , Immunity, Cellular
15.
Front Immunol ; 12: 780810, 2021.
Article in English | MEDLINE | ID: mdl-34899745

ABSTRACT

Background: Trypanosomatids are protozoa responsible for a wide range of diseases, with emphasis on Chagas Disease (CD) and Leishmaniasis, which are in the list of most relevant Neglected Tropical Diseases (NTD) according to World Health Organization (WHO). During the infectious process, immune system is immediately activated, and parasites can invade nucleated cells through a broad diversity of receptors. The complement system - through classical, alternative and lectin pathways - plays a role in the first line of defense against these pathogens, acting in opsonization, phagocytosis and lysis of parasites. Genetic modifications in complement genes, such as Single Nucleotide Polymorphisms (SNPs), can influence host susceptibility to these parasites and modulate protein expression. Methods: In March and April 2021, a literature search was conducted at the PubMed and Google Scholar databases and the reference lists obtained were verified. After applying the inclusion and exclusion criteria, the selected studies were evaluated and scored according to eleven established criteria regarding their thematic approach and design, aiming at the good quality of publications. Results: Twelve papers were included in this systematic review: seven investigating CD and five focusing on Leishmaniasis. Most articles presented gene and protein approaches, careful determination of experimental groups, and adequate choice of experimental techniques, although several of them were not up-to-date. Ten studies explored the association of polymorphisms and haplotypes with disease progression, with emphasis on lectin complement pathway genes. Decreased and increased patient serum protein levels were associated with susceptibility to CD and Visceral Leishmaniasis, respectively. Conclusion: This systematic review shows the influence of genetic alterations in complement genes on the progression of several infectious diseases, with a focus on conditions caused by trypanosomatids, and contributes suggestions and evidence to improve experimental design in future research proposals.


Subject(s)
Chagas Disease/parasitology , Complement Activation/genetics , Complement System Proteins/genetics , Genetic Variation , Leishmania/pathogenicity , Leishmaniasis/parasitology , Trypanosoma cruzi/pathogenicity , Chagas Disease/genetics , Chagas Disease/immunology , Chagas Disease/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Progression , Genetic Predisposition to Disease , Host-Parasite Interactions , Humans , Leishmania/immunology , Leishmaniasis/genetics , Leishmaniasis/immunology , Leishmaniasis/metabolism , Phenotype , Risk Assessment , Risk Factors , Trypanosoma cruzi/immunology
16.
PLoS One ; 16(12): e0262158, 2021.
Article in English | MEDLINE | ID: mdl-34972189

ABSTRACT

Leishmaniasis is a disease caused by the protozoan parasite Leishmania and is known to affect millions of individuals worldwide. In recent years, we have established the critical role played by Leishmania zinc-metalloprotease GP63 in the modulation of host macrophage signalling and functions, favouring its survival and progression within its host. Leishmania major lacking GP63 was reported to cause limited infection in mice, however, it is still unclear how GP63 may influence the innate inflammatory response and parasite survival in an in vivo context. Therefore, we were interested in analyzing the early innate inflammatory events upon Leishmania inoculation within mice and establish whether Leishmania GP63 influences this initial inflammatory response. Experimentally, L. major WT (L. majorWT), L. major GP63 knockout (L. majorKO), or L. major GP63 rescue (L. majorR) were intraperitoneally inoculated in mice and the inflammatory cells recruited were characterized microscopically and by flow cytometry (number and cell type), and their infection determined. Pro-inflammatory markers such as cytokines, chemokines, and extracellular vesicles (EVs, e.g. exosomes) were monitored and proteomic analysis was performed on exosome contents. Data obtained from this study suggest that Leishmania GP63 does not significantly influence the pathogen-induced inflammatory cell recruitment, but rather their activation status and effector function. Concordantly, internalization of promastigotes during early infection could be influenced by GP63 as fewer L. majorKO amastigotes were found within host cells and appear to maintain in host cells over time. Collectively this study provides a clear analysis of innate inflammatory events occurring during L. major infection and further establish the prominent role of the virulence factor GP63 to provide favourable conditions for host cell infection.


Subject(s)
Leishmania major/metabolism , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis/immunology , Leishmaniasis/parasitology , Metalloendopeptidases/chemistry , Animals , Computational Biology , Exosomes/metabolism , Female , Host-Parasite Interactions/physiology , Inflammation/immunology , Inflammation/metabolism , Leishmania , Metalloproteases/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Nanoparticles/chemistry , Proteomics/methods , RNA-Seq
17.
Front Immunol ; 12: 750648, 2021.
Article in English | MEDLINE | ID: mdl-34790196

ABSTRACT

Background: Leishmaniasis is a neglected arthropod-borne disease that affects millions of people worldwide. Successful Leishmania infections require the mitigation of immune cell functions leading to parasite survival and proliferation. A large body of evidence highlights the involvement of neutrophils (PMNs) and dendritic cells (DCs) in the establishment of immunological responses against these parasites. However, few studies, contemplate to what extent these cells interact synergistically to constrain Leishmania infection. Objective: We sought to investigate how PMNs and infected DCs interact in an in vitro model of Leishmania amazonensis infection. Material and Methods: Briefly, human PMNs and DCs were purified from the peripheral blood of healthy donors. Next, PMNs were activated with fibronectin and subsequently co-cultured with L. amazonensis-infected DCs. Results: We observed that L. amazonensis-infected DC exhibited lower rates of infection when co-cultivated with either resting or activated PMNs. Surprisingly, we found that the release of neutrophil enzymes was not involved in Leishmania killing. Next, we showed that the interaction between PMNs and infected-DCs was intermediated by DC-SIGN, further suggesting that parasite elimination occurs in a contact-dependent manner. Furthermore, we also observed that TNFα and ROS production was dependent on DC-SIGN-mediated contact, as well as parasite elimination is dependent on TNFα production in the co-culture. Finally, we observed that direct contact between PMNs and DCs are required to restore the expression of DC maturation molecules during L. amazonensis infection. Conclusion: Our findings suggest that the engagement of direct contact between PMNs and L. amazonensis-infected DC via DC-SIGN is required for the production of inflammatory mediators with subsequent parasite elimination and DC maturation.


Subject(s)
Cell Adhesion Molecules/immunology , Dendritic Cells/immunology , Lectins, C-Type/immunology , Leishmaniasis/immunology , Neutrophils/immunology , Receptors, Cell Surface/immunology , Cell Differentiation , Cells, Cultured , Coculture Techniques , Humans , Leishmania , Leishmaniasis/parasitology , Tumor Necrosis Factor-alpha/immunology
18.
Immunity ; 54(12): 2724-2739.e10, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34687607

ABSTRACT

Nitric oxide (NO) is an important antimicrobial effector but also prevents unnecessary tissue damage by shutting down the recruitment of monocyte-derived phagocytes. Intracellular pathogens such as Leishmania major can hijack these cells as a niche for replication. Thus, NO might exert containment by restricting the availability of the cellular niche required for efficient pathogen proliferation. However, such indirect modes of action remain to be established. By combining mathematical modeling with intravital 2-photon biosensors of pathogen viability and proliferation, we show that low L. major proliferation results not from direct NO impact on the pathogen but from reduced availability of proliferation-permissive host cells. Although inhibiting NO production increases recruitment of these cells, and thus pathogen proliferation, blocking cell recruitment uncouples the NO effect from pathogen proliferation. Therefore, NO fulfills two distinct functions for L. major containment: permitting direct killing and restricting the supply of proliferation-permissive host cells.


Subject(s)
Leishmania major/physiology , Leishmaniasis/immunology , Macrophages/immunology , Nitric Oxide/metabolism , Animals , Cell Growth Processes , Cell Movement , Cell Proliferation , Disease Models, Animal , Host-Pathogen Interactions , Humans , Intravital Microscopy , Mice , Mice, Inbred C57BL , Models, Theoretical
19.
Parasit Vectors ; 14(1): 525, 2021 Oct 10.
Article in English | MEDLINE | ID: mdl-34629081

ABSTRACT

BACKGROUND: Immunotherapeutic drugs, such as domperidone, have been shown to be promising treatments against canine leishmaniosis (CanL), but limited data are available. The aim of this pilot study (therapeutic, prospective and non-controlled) was to evaluate the effect of domperidone on serum antibody titers of Leishmania infantum, globulins, gamma globulins, acute-phase proteins (e.g. C-reactive protein [CRP]), big endothelin-1 (big ET-1), serum creatinine (SC) and proteinuria in dogs with leishmaniosis affected by chronic kidney disease (CKD). METHODS: Dogs were recruited if "exposed" to or "infected" with L. infantum and affected by CKD (IRIS stage 1 [proteinuric] or IRIS stage 2-3a [SC < 3.5 mg/dl; proteinuric or non-proteinuric]). After inclusion, an oral suspension of domperidone was administered, and the dogs were followed up for 180 days, with checks at 30, 60, 90 and 180 days after initial treatment. RESULTS: Of the 14 recruited dogs, nine showed a statistically significant reduction in SC (χ2 = 9.1, df = 3, P = 0.028), but not in the urine protein/creatinine ratio (χ2 = 6.43, df = 3, P = 0.092). All dogs showed a significant reduction in antibody titers for L. infantum (χ2 = 9.56, df = 2, P = 0.008), globulins (χ2 = 11.08, df = 3, P = 0.011) and gamma globulins (χ2 = 12.38, df = 3, P = 0.006) during the study period. There was also a statistically significant reduction in CRP (χ2 = 16.7, df = 3, P = 0.001), but not in big ET-1 (χ2 = 2.04, df = 3, P = 0.563). CONCLUSIONS: This study provides preliminary results on the ability of domperidone to improve SC and reduce anti-L. infantum antibody titers, globulins, gamma globulins and CRP in dogs with leishmaniosis and CKD.


Subject(s)
Antibodies, Protozoan/blood , Creatinine/blood , Domperidone/therapeutic use , Inflammation/blood , Leishmania infantum/immunology , Leishmaniasis/drug therapy , Leishmaniasis/veterinary , Renal Insufficiency, Chronic/drug therapy , Acute-Phase Proteins , Animals , Biomarkers/blood , Dog Diseases/drug therapy , Dog Diseases/immunology , Dog Diseases/parasitology , Dogs , Female , Leishmania infantum/drug effects , Leishmaniasis/immunology , Male , Pilot Projects , Prospective Studies , Renal Insufficiency, Chronic/blood
20.
PLoS Negl Trop Dis ; 15(8): e0009681, 2021 08.
Article in English | MEDLINE | ID: mdl-34398874

ABSTRACT

Vitamin D (VitD) deficiency has been shown to be a risk factor for a plethora of disorders. We have shown that dogs with clinical leishmaniasis presented lower VitD serum levels than non-infected dogs, and even lower than those with asymptomatic infection. However, if VitD deficiency is a risk factor to develop clinical leishmaniasis remains to be answered. It is also unknown if VitD participates in Leishmania control. First, we retrospectively analysed VitD concentration in serum samples from 36 healthy dogs collected in different periods of the year concluding that there isn't a seasonal variation of this vitamin in dogs. We also included 9 dogs with clinical leishmaniasis and 10 non-infected healthy dogs, in which we measured VitD levels at the beginning of the study, when all dogs were negative for serology and qPCR, and 1 year later. Whereas non-infected dogs showed no change in VitD levels along the study, those developing clinical leishmaniasis showed a significant VitD reduction at the end of the study (35%). When we compared VitD concentration between the two groups at the beginning of the study, no differences were detected (43.6 (38-59) ng/mL, P = 0.962). Furthermore, an in vitro model using a canine macrophage cell line proved that adding active VitD leads to a significant reduction in L. infantum load (31.4%). Analyzing expression of genes related to VitD pathway on primary canine monocytes, we showed that CBD103 expression was significantly enhanced after 1,25(OH)2D addition. Our results show that VitD concentration is neither seasonal nor a risk factor for developing canine leishmaniasis, but it diminishes with the onset of clinical disease suggesting a role in parasitic control. Our in vitro results corroborate this hypothesis and point out that VitD regulates infection through CBD103 expression. These results open the possibility for studies testing VitD as an adjuvant in leishmaniasis therapy.


Subject(s)
Dog Diseases/immunology , Leishmaniasis/veterinary , Vitamin D/blood , beta-Defensins/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Dog Diseases/blood , Dog Diseases/drug therapy , Dog Diseases/genetics , Dogs , Female , Leishmania infantum/physiology , Leishmaniasis/blood , Leishmaniasis/drug therapy , Leishmaniasis/immunology , Male , Monocytes/immunology , Retrospective Studies , Seasons , Vitamin D/administration & dosage , beta-Defensins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL