Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.816
1.
Parasit Vectors ; 17(1): 203, 2024 May 07.
Article En | MEDLINE | ID: mdl-38711063

BACKGROUND: The role of pathogen genotype in determining disease severity and immunopathology has been studied intensively in microbial pathogens including bacteria, fungi, protozoa and viruses but is poorly understood in parasitic helminths. The medically important blood fluke Schistosoma mansoni is an excellent model system to study the impact of helminth genetic variation on immunopathology. Our laboratory has demonstrated that laboratory schistosome populations differ in sporocyst growth and cercarial production in the intermediate snail host and worm establishment and fecundity in the vertebrate host. Here, we (i) investigate the hypothesis that schistosome genotype plays a significant role in immunopathology and related parasite life history traits in the vertebrate mouse host and (ii) quantify the relative impact of parasite and host genetics on infection outcomes. METHODS: We infected BALB/c and C57BL/6 mice with four different laboratory schistosome populations from Africa and the Americas. We quantified disease progression in the vertebrate host by measuring body weight and complete blood count (CBC) with differential over a 12-week infection period. On sacrifice, we assessed parasitological (egg and worm counts, fecundity), immunopathological (organ measurements and histopathology) and immunological (CBC with differential and cytokine profiles) characteristics to determine the impact of parasite and host genetics. RESULTS: We found significant variation between parasite populations in worm numbers, fecundity, liver and intestine egg counts, liver and spleen weight, and fibrotic area but not in granuloma size. Variation in organ weight was explained by egg burden and intrinsic parasite factors independent of egg burden. We found significant variation between infected mouse lines in cytokine levels (IFN-γ, TNF-α), eosinophils, lymphocytes and monocyte counts. CONCLUSIONS: This study showed that both parasite and host genotype impact the outcome of infection. While host genotype explains most of the variation in immunological traits, parasite genotype explains most of the variation in parasitological traits, and both host and parasite genotypes impact immunopathology outcomes.


Genotype , Mice, Inbred BALB C , Mice, Inbred C57BL , Schistosoma mansoni , Schistosomiasis mansoni , Animals , Schistosoma mansoni/immunology , Schistosoma mansoni/genetics , Mice , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Schistosomiasis mansoni/pathology , Female , Host-Parasite Interactions/immunology , Host-Parasite Interactions/genetics , Cytokines/genetics , Cytokines/blood , Cytokines/immunology
2.
Front Immunol ; 15: 1342431, 2024.
Article En | MEDLINE | ID: mdl-38655255

Chagas disease, caused by Trypanosoma cruzi, remains a serious public health problem worldwide. The parasite was subdivided into six distinct genetic groups, called "discrete typing units" (DTUs), from TcI to TcVI. Several studies have indicated that the heterogeneity of T. cruzi species directly affects the diversity of clinical manifestations of Chagas disease, control, diagnosis performance, and susceptibility to treatment. Thus, this review aims to describe how T. cruzi genetic diversity influences the biology of the parasite and/or clinical parameters in humans. Regarding the geographic dispersion of T. cruzi, evident differences were observed in the distribution of DTUs in distinct areas. For example, TcII is the main DTU detected in Brazilian patients from the central and southeastern regions, where there are also registers of TcVI as a secondary T. cruzi DTU. An important aspect observed in previous studies is that the genetic variability of T. cruzi can impact parasite infectivity, reproduction, and differentiation in the vectors. It has been proposed that T. cruzi DTU influences the host immune response and affects disease progression. Genetic aspects of the parasite play an important role in determining which host tissues will be infected, thus heavily influencing Chagas disease's pathogenesis. Several teams have investigated the correlation between T. cruzi DTU and the reactivation of Chagas disease. In agreement with these data, it is reasonable to suppose that the immunological condition of the patient, whether or not associated with the reactivation of the T. cruzi infection and the parasite strain, may have an important role in the pathogenesis of Chagas disease. In this context, understanding the genetics of T. cruzi and its biological and clinical implications will provide new knowledge that may contribute to additional strategies in the diagnosis and clinical outcome follow-up of patients with Chagas disease, in addition to the reactivation of immunocompromised patients infected with T. cruzi.


Chagas Disease , Genetic Variation , Trypanosoma cruzi , Trypanosoma cruzi/genetics , Humans , Chagas Disease/immunology , Chagas Disease/parasitology , Animals , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology
3.
BMC Biol ; 22(1): 89, 2024 Apr 22.
Article En | MEDLINE | ID: mdl-38644510

BACKGROUND: Innate immune responses can be activated by pathogen-associated molecular patterns (PAMPs), danger signals released by damaged tissues, or the absence of self-molecules that inhibit immunity. As PAMPs are typically conserved across broad groups of pathogens but absent from the host, it is unclear whether they allow hosts to recognize parasites that are phylogenetically similar to themselves, such as parasitoid wasps infecting insects. RESULTS: Parasitoids must penetrate the cuticle of Drosophila larvae to inject their eggs. In line with previous results, we found that the danger signal of wounding triggers the differentiation of specialized immune cells called lamellocytes. However, using oil droplets to mimic infection by a parasitoid wasp egg, we found that this does not activate the melanization response. This aspect of the immune response also requires exposure to parasite molecules. The unidentified factor enhances the transcriptional response in hemocytes and induces a specific response in the fat body. CONCLUSIONS: We conclude that a combination of danger signals and the recognition of nonself molecules is required to activate Drosophila's immune response against parasitic insects.


Hemocytes , Host-Parasite Interactions , Immunity, Innate , Wasps , Animals , Wasps/physiology , Host-Parasite Interactions/immunology , Hemocytes/immunology , Drosophila melanogaster/parasitology , Drosophila melanogaster/immunology , Drosophila melanogaster/physiology , Larva/immunology , Larva/parasitology , Drosophila/parasitology , Drosophila/immunology
4.
Bull Math Biol ; 86(6): 62, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38662120

Hosts can evolve a variety of defences against parasitism, including resistance (which prevents or reduces the spread of infection) and tolerance (which protects against virulence). Some organisms have evolved different levels of tolerance at different life-stages, which is likely to be the result of coevolution with pathogens, and yet it is currently unclear how coevolution drives patterns of age-specific tolerance. Here, we use a model of tolerance-virulence coevolution to investigate how age structure influences coevolutionary dynamics. Specifically, we explore how coevolution unfolds when tolerance and virulence (disease-induced mortality) are age-specific compared to when these traits are uniform across the host lifespan. We find that coevolutionary cycling is relatively common when host tolerance is age-specific, but cycling does not occur when tolerance is the same across all ages. We also find that age-structured tolerance can lead to selection for higher virulence in shorter-lived than in longer-lived hosts, whereas non-age-structured tolerance always leads virulence to increase with host lifespan. Our findings therefore suggest that age structure can have substantial qualitative impacts on host-pathogen coevolution.


Biological Evolution , Host-Pathogen Interactions , Mathematical Concepts , Virulence , Animals , Age Factors , Models, Biological , Host-Parasite Interactions/immunology , Biological Coevolution , Humans , Longevity
5.
Trends Parasitol ; 40(5): 386-400, 2024 May.
Article En | MEDLINE | ID: mdl-38609741

Obesity is a worldwide pandemic and major risk factor for the development of metabolic syndrome (MetS) and type 2 diabetes (T2D). T2D requires lifelong medical support to limit complications and is defined by impaired glucose tolerance, insulin resistance (IR), and chronic low-level systemic inflammation initiating from adipose tissue. The current preventative strategies include a healthy diet, controlled physical activity, and medication targeting hyperglycemia, with underexplored underlying inflammation. Studies suggest a protective role for helminth infection in the prevention of T2D. The mechanisms may involve induction of modified type 2 and regulatory immune responses that suppress inflammation and promote insulin sensitivity. In this review, the roles of helminths in counteracting MetS, and prospects for harnessing these protective mechanisms for the development of novel anti-diabetes drugs are discussed.


Diabetes Mellitus, Type 2 , Helminths , Metabolic Syndrome , Animals , Humans , Helminths/immunology , Helminths/physiology , Metabolic Syndrome/immunology , Metabolic Syndrome/metabolism , Metabolic Syndrome/parasitology , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Helminthiasis/immunology , Helminthiasis/parasitology , Obesity/immunology , Obesity/metabolism , Host-Parasite Interactions/immunology , Insulin Resistance
6.
Nature ; 623(7985): 149-156, 2023 Nov.
Article En | MEDLINE | ID: mdl-37880367

Host factors that mediate Leishmania genetic exchange are not well defined. Here we demonstrate that natural IgM (IgMn)1-4 antibodies mediate parasite genetic exchange by inducing the transient formation of a spherical parasite clump that promotes parasite fusion and hybrid formation. We establish that IgMn from Leishmania-free animals binds to the surface of Leishmania parasites to induce significant changes in the expression of parasite transcripts and proteins. Leishmania binding to IgMn is partially lost after glycosidase treatment, although parasite surface phosphoglycans, including lipophosphoglycan, are not required for IgMn-induced parasite clumping. Notably, the transient formation of parasite clumps is essential for Leishmania hybridization in vitro. In vivo, we observed a 12-fold increase in hybrid formation in sand flies provided a second blood meal containing IgMn compared with controls. Furthermore, the generation of recombinant progeny from mating hybrids and parental lines were only observed in sand flies provided with IgMn. Both in vitro and in vivo IgM-induced Leishmania crosses resulted in full genome hybrids that show equal patterns of biparental contribution. Leishmania co-option of a host natural antibody to facilitate mating in the insect vector establishes a new paradigm of parasite-host-vector interdependence that contributes to parasite diversity and fitness by promoting genetic exchange.


Host-Parasite Interactions , Immunoglobulin M , Leishmania , Psychodidae , Reproduction , Animals , Hybridization, Genetic , Immunoglobulin M/immunology , Leishmania/genetics , Leishmania/immunology , Psychodidae/immunology , Psychodidae/parasitology , Reproduction/genetics , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology , Gene Expression Regulation , Glycoside Hydrolases/metabolism
7.
Science ; 379(6628): eabl3837, 2023 01 13.
Article En | MEDLINE | ID: mdl-36634189

Ancestral signaling pathways serve critical roles in metazoan development, physiology, and immunity. We report an evolutionary interspecies communication pathway involving a central Ixodes scapularis tick receptor termed Dome1, which acquired a mammalian cytokine receptor motif exhibiting high affinity for interferon-gamma (IFN-γ). Host-derived IFN-γ facilitates Dome1-mediated activation of the Ixodes JAK-STAT pathway. This accelerates tick blood meal acquisition and development while upregulating antimicrobial components. The Dome1-JAK-STAT pathway, which exists in most Ixodid tick genomes, regulates the regeneration and proliferation of gut cells-including stem cells-and dictates metamorphosis through the Hedgehog and Notch-Delta networks, ultimately affecting Ixodes vectorial competence. We highlight the evolutionary dependence of I. scapularis on mammalian hosts through cross-species signaling mechanisms that dually influence arthropod immunity and development.


Arachnid Vectors , Host-Parasite Interactions , Ixodes , Janus Kinases , Receptors, Cytokine , STAT Transcription Factors , Animals , Interferon-gamma/metabolism , Ixodes/genetics , Ixodes/immunology , Janus Kinases/genetics , Janus Kinases/metabolism , Signal Transduction , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Host-Parasite Interactions/immunology , Receptors, Cytokine/metabolism , Arachnid Vectors/immunology
8.
Parasit Vectors ; 15(1): 454, 2022 Dec 05.
Article En | MEDLINE | ID: mdl-36471417

BACKGROUND: Toxoplasma gondii is known as the most successful parasite, which can regulate the host immune response through a variety of ways to achieve immune escape. We previously reported that a novel gene wx2 of T. gondii may be a virulence-related molecule. The objective of this study was to explore the mechanism of wx2 regulating host immune response. METHODS: The wx2 knockout strain (RHwx2-/- strain) and complementary strain (RHwx2+/+ strain) were constructed by the CRISPR/Cas9 technique, and the virulence of the wx2 gene was detected and changes in pyroptosis-related molecules were observed. RESULTS: Compared with the wild RH and RHwx2+/+ strain groups, the survival time for mice infected with the RHwx2-/- strain was prolonged to a certain extent. The mRNA levels of pyroptosis-related molecules of caspase-1, NLRP3, and GSDMD and et al. in mouse lymphocytes in vivo and RAW267.4 cells in vitro infected with RHwx2-/- strain increased to different degrees, compared with infected with wild RH strain and RHwx2+/+ strain. As with the mRNA level, the protein level of caspase-1, caspase-1 p20, IL-1ß, NLRP3, GSDMD-FL, GSDMD-N, and phosphorylation level of NF-κB (p65) were also significantly increased. These data suggest that wx2 may regulate the host immune response through the pyroptosis pathway. In infected RAW264.7 cells at 48 h post-infection, the levels of Th1-type cytokines of IFN-γ, Th2-type cytokines such as IL-13, Th17-type cytokine of IL-17 in cells infected with RHwx2-/- were significantly higher than those of RH and RHwx2+/+ strains, suggesting that the wx2 may inhibit the host's immune response. CONCLUSION: wx2 is a virulence related gene of T. gondii, and may be involved in host immune regulation by inhibiting the pyroptosis pathway.


Host-Parasite Interactions , Pyroptosis , Toxoplasma , Animals , Mice , Caspase 1/metabolism , Cytokines/genetics , Cytokines/metabolism , Immunity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , RNA, Messenger/metabolism , Toxoplasma/genetics , Toxoplasma/pathogenicity , Virulence , Host-Parasite Interactions/immunology
9.
Nature ; 611(7936): 563-569, 2022 Nov.
Article En | MEDLINE | ID: mdl-36352220

Malaria infection involves an obligatory, yet clinically silent liver stage1,2. Hepatocytes operate in repeating units termed lobules, exhibiting heterogeneous gene expression patterns along the lobule axis3, but the effects of hepatocyte zonation on parasite development at the molecular level remain unknown. Here we combine single-cell RNA sequencing4 and single-molecule transcript imaging5 to characterize the host and parasite temporal expression programmes in a zonally controlled manner for the rodent malaria parasite Plasmodium berghei ANKA. We identify differences in parasite gene expression in distinct zones, including potentially co-adaptive programmes related to iron and fatty acid metabolism. We find that parasites develop more rapidly in the pericentral lobule zones and identify a subpopulation of periportally biased hepatocytes that harbour abortive infections, reduced levels of Plasmodium transcripts and parasitophorous vacuole breakdown. These 'abortive hepatocytes', which appear predominantly with high parasite inoculum, upregulate immune recruitment and key signalling programmes. Our study provides a resource for understanding the liver stage of Plasmodium infection at high spatial resolution and highlights the heterogeneous behaviour of both the parasite and the host hepatocyte.


Gene Expression Regulation , Hepatocytes , Liver , Malaria , Parasites , Plasmodium berghei , Single-Cell Analysis , Animals , Hepatocytes/cytology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/parasitology , Liver/anatomy & histology , Liver/cytology , Liver/immunology , Liver/parasitology , Malaria/genetics , Malaria/immunology , Malaria/parasitology , Parasites/genetics , Parasites/immunology , Parasites/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Plasmodium berghei/metabolism , Single Molecule Imaging , Sequence Analysis, RNA , Iron/metabolism , Fatty Acids/metabolism , Transcription, Genetic , Genes, Protozoan/genetics , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology
10.
Nat Commun ; 13(1): 977, 2022 02 21.
Article En | MEDLINE | ID: mdl-35190553

Some snails act as intermediate hosts (vectors) for parasitic flatworms (flukes) that cause neglected tropical diseases, such as schistosomiases. Schistosoma haematobium is a blood fluke that causes urogenital schistosomiasis and induces bladder cancer and increased risk of HIV infection. Understanding the molecular biology of the snail and its relationship with the parasite could guide development of an intervention approach that interrupts transmission. Here, we define the genome for a key intermediate host of S. haematobium-called Bulinus truncatus-and explore protein groups inferred to play an integral role in the snail's biology and its relationship with the schistosome parasite. Bu. truncatus shared many orthologous protein groups with Biomphalaria glabrata-the key snail vector for S. mansoni which causes hepatointestinal schistosomiasis in people. Conspicuous were expansions in signalling and membrane trafficking proteins, peptidases and their inhibitors as well as gene families linked to immune response regulation, such as a large repertoire of lectin-like molecules. This work provides a sound basis for further studies of snail-parasite interactions in the search for targets to block schistosomiasis transmission.


Bulinus/genetics , Cell Nucleus/genetics , Disease Vectors , Schistosomiasis haematobia/transmission , Animals , Bulinus/parasitology , Genome , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology , Humans , Schistosoma haematobium/immunology , Schistosomiasis haematobia/parasitology
11.
Front Immunol ; 13: 801120, 2022.
Article En | MEDLINE | ID: mdl-35154114

An increase in mast cells (MCs) and MCs mediators has been observed in malaria-associated bacteremia, however, the role of these granulocytes in malarial immunity is poorly understood. Herein, we studied the role of mouse MC protease (Mcpt) 4, an ortholog of human MC chymase, in malaria-induced bacteremia using Mcpt4 knockout (Mcpt4-/-) mice and Mcpt4+/+ C57BL/6J controls, and the non-lethal mouse parasite Plasmodium yoelii yoelii 17XNL. Significantly lower parasitemia was observed in Mcpt4-/- mice compared with Mcpt4+/+ controls by day 10 post infection (PI). Although bacterial 16S DNA levels in blood were not different between groups, increased intestinal permeability to FITC-dextran and altered ileal adherens junction E-cadherin were observed in Mcpt4-/- mice. Relative to infected Mcpt4+/+ mice, ileal MC accumulation in Mcpt4-/- mice occurred two days earlier and IgE levels were higher by days 8-10 PI. Increased levels of circulating myeloperoxidase were observed at 6 and 10 days PI in Mcpt4+/+ but not Mcpt4-/- mice, affirming a role for neutrophil activation that was not predictive of parasitemia or bacterial 16S copies in blood. In contrast, early increased plasma levels of TNF-α, IL-12p40 and IL-3 were observed in Mcpt4-/- mice, while levels of IL-2, IL-10 and MIP1ß (CCL4) were increased over the same period in Mcpt4+/+ mice, suggesting that the host response to infection was skewed toward a type-1 immune response in Mcpt4-/- mice and type-2 response in Mcpt4+/+ mice. Spearman analysis revealed an early (day 4 PI) correlation of Mcpt4-/- parasitemia with TNF-α and IFN-γ, inflammatory cytokines known for their roles in pathogen clearance, a pattern that was observed in Mcpt4+/+ mice much later (day 10 PI). Transmission success of P. y. yoelii 17XNL to Anopheles stephensi was significantly higher from infected Mcpt4-/- mice compared with infected Mcpt4+/+ mice, suggesting that Mcpt4 also impacts transmissibility of sexual stage parasites. Together, these results suggest that early MCs activation and release of Mcpt4 suppresses the host immune response to P. y. yoelii 17XNL, perhaps via degradation of TNF-α and promotion of a type-2 immune response that concordantly protects epithelial barrier integrity, while limiting the systemic response to bacteremia and parasite transmissibility.


Anopheles/parasitology , Cell Membrane Permeability/immunology , Chymases/genetics , Chymases/immunology , Host-Parasite Interactions/immunology , Malaria/immunology , Mast Cells/enzymology , Plasmodium yoelii/immunology , Animals , Female , Ileum/cytology , Ileum/pathology , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism
12.
PLoS One ; 17(2): e0263517, 2022.
Article En | MEDLINE | ID: mdl-35180232

Endoparasitism is a common disease in dogs throughout their lifetime despite the widespread availability of inexpensive diagnostic tests and effective treatments. The consequences of host parasite interactions in otherwise apparently healthy dogs remains largely unknown. This cross-sectional study used complete blood count, serum biochemistry, and fecal flotation data collected from 3,018 young dogs (<3 years of age) enrolled within the Morris Animal Foundation Golden Retriever Lifetime Study (GRLS) to determine the prevalence of endoparasitism and compare bloodwork values of parasite positive and negative participants using logistic regression. Variables including age, gender, reproductive status, and geographic region at the time of evaluation were assessed to identify potential associations. To the authors' knowledge, a comprehensive assessment of clinicopathological changes associated with endoparasite infection in a large cohort has not been completed in the recent decade. The overall prevalence of endoparasitism was 6.99% (211/3018). Dogs who were parasite positive had statistically lower albumin (P = 0.004), lower RBC count (P = 0.01), higher neutrophil count (P = 0.002), and higher platelet count (P <0.001) as compared to parasite negative dogs. It was also concluded that dogs living in rural areas were more likely to have endoparasites than those living in suburban areas. Epidemiological data is crucial for the design and monitoring of prevention and control strategies. Identification of endoparasites by fecal testing is an essential tool to identify susceptible and resistant animals that can act as spreaders and reservoirs of intestinal parasites thereby enabling appropriate therapy and reducing the risk of new infection to animals and humans. Further epidemiological studies are needed to prevent, monitor, and develop new strategies to control endoparasites.


Dog Diseases/blood , Dog Diseases/epidemiology , Helminths/isolation & purification , Intestinal Diseases, Parasitic/epidemiology , Intestinal Diseases, Parasitic/veterinary , Parasites/isolation & purification , Animals , Cohort Studies , Cross-Sectional Studies , Dog Diseases/diagnosis , Dog Diseases/parasitology , Dogs , Feces/parasitology , Female , Helminths/classification , Host-Parasite Interactions/immunology , Intestinal Diseases, Parasitic/blood , Intestinal Diseases, Parasitic/diagnosis , Leukocyte Count , Logistic Models , Male , Neutrophils/immunology , Odds Ratio , Parasites/classification , Platelet Count , Prevalence , Risk Factors
13.
Infect Immun ; 90(1): e0037721, 2022 01 25.
Article En | MEDLINE | ID: mdl-34694918

Plasmodium falciparum cysteine-rich protective antigen (CyRPA) is a conserved component of an essential erythrocyte invasion complex (RH5/Ripr/CyRPA) and a target of potent cross-strain parasite-neutralizing antibodies. While naturally acquired human RH5 antibodies have been functionally characterized, there are no similar reports on CyRPA. Thus, we analyzed the parasite-neutralizing activity of naturally acquired human CyRPA antibodies. In this regard, CyRPA human antibodies were measured and purified from malaria-infected plasma obtained from patients in central India and analyzed for their parasite neutralizing activity via in vitro growth inhibition assays (GIA). We report that, despite being susceptible to antibodies, CyRPA is a highly conserved antigen that does not appear to be under substantial immune selection pressure, as a very low acquisition rate for anti-CyRPA antibodies was reported in malaria-exposed Indians. We demonstrate for the first time that the small amounts of natural CyRPA antibodies exhibited functional parasite-neutralizing activity and that a CyRPA-based vaccine formulation induces highly potent antibodies in rabbits. Importantly, the vaccine-induced CyRPA antibodies exhibited a robust 50% inhibitory concentration (IC50) of 21.96 µg/ml, which is comparable to the IC50 of antibodies against the leading blood-stage vaccine candidate, reticulocyte-binding-like homologous protein 5 (RH5). Our data support CyRPA as a unique vaccine target that is highly susceptible to immune attack but is highly conserved compared to other leading candidates such as MSP-1 and AMA-1, further substantiating its promise as a leading blood-stage vaccine candidate.


Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Host-Parasite Interactions/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Antibodies, Neutralizing/immunology , Antibody Specificity/immunology , Disease Resistance/immunology , Enzyme-Linked Immunosorbent Assay , Erythrocytes/immunology , Erythrocytes/parasitology , Humans , Malaria Vaccines/immunology , Malaria, Falciparum/parasitology , Recombinant Proteins/immunology
14.
Int Immunopharmacol ; 102: 108400, 2022 Jan.
Article En | MEDLINE | ID: mdl-34890999

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.


Leishmania/immunology , Leishmaniasis/immunology , Animals , Host-Parasite Interactions/immunology , Humans , Immunity, Cellular
15.
Immunology ; 165(2): 219-233, 2022 02.
Article En | MEDLINE | ID: mdl-34775598

Tuberculosis (TB) and malaria remain serious threats to global health. Bacillus Calmette-Guerin (BCG), the only licensed vaccine against TB protects against severe disseminated forms of TB in infants but shows poor efficacy against pulmonary TB in adults. Co-infections have been reported as one of the factors implicated in vaccine inefficacy. Given the geographical overlap of malaria and TB in areas where BCG vaccination is routinely administered, we hypothesized that virulence-dependent co-infection with Plasmodium species could alter the BCG-specific immune responses thus resulting in failure to protect against Mycobacterium tuberculosis. We compared virulent Plasmodium berghei and non-virulent Plasmodium chabaudi, their effects on B cells, effector and memory T cells, and the outcome on BCG-induced efficacy against M. tuberculosis infection. We demonstrate that malaria co-infection modulates both B- and T-cell immune responses but does not significantly alter the ability of the BCG vaccine to inhibit the growth of M. tuberculosis irrespective of parasite virulence. This malaria-driven immune regulation may have serious consequences in the early clinical trials of novel vaccines, which rely on vaccine-specific T-cell responses to screen novel vaccines for progression to the more costly vaccine efficacy trials.


BCG Vaccine/immunology , Host-Parasite Interactions/immunology , Host-Pathogen Interactions/immunology , Immunogenicity, Vaccine , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/prevention & control , Tuberculosis/prevention & control , Animals , Apoptosis , CD4 Lymphocyte Count , Disease Models, Animal , Female , Humans , Malaria/immunology , Malaria/parasitology , Memory T Cells/immunology , Memory T Cells/metabolism , Mice , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tuberculosis Vaccines/immunology , Vaccination
16.
Cells ; 10(12)2021 12 06.
Article En | MEDLINE | ID: mdl-34943932

The extracellular protozoan parasite Giardia duodenalis is a well-known and important causative agent of diarrhea on a global scale. Macrophage pyroptosis has been recognized as an important innate immune effector mechanism against intracellular pathogens. Yet, the effects of noninvasive Giardia infection on macrophage pyroptosis and the associated molecular triggers and regulators remain poorly defined. Here we initially observed that NLRP3 inflammasome-mediated pyroptosis was activated in Giardia-treated macrophages, and inhibition of ROS, NLRP3, or caspase-1 could block GSDMD cleavage, IL-1ß, IL-18 and LDH release, and the cell viability reduction. We also confirmed that Giardia-induced NLRP3 inflammasome activation was involved in its K63 deubiquitination. Thus, six candidate deubiquitinases were screened, among which A20 was identified as an effective regulator. We then screened TLRs on macrophage membranes and found that upon stimulation TLR4 was tightly correlated to ROS enhancement, A20-mediated NLRP3 deubiquitination, and pyroptotic signaling. In addition, several Giardia-secreted proteins were predicted as trigger factors via secretome analysis, of which peptidyl-prolyl cis-trans isomerase B (PPIB) independently induced macrophage pyroptosis. This was similar to the findings from the trophozoite treatment, and also led to the TLR4-mediated activation of NLRP3 through K63 deubiquitination by A20. Collectively, the results of this study have significant implications for expanding our understanding of host defense mechanisms after infection with G. duodenalis.


Diarrhea/genetics , Giardia lamblia/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Animals , Cell Survival/drug effects , Deubiquitinating Enzymes/genetics , Diarrhea/immunology , Diarrhea/parasitology , Disease Models, Animal , Giardia lamblia/immunology , Giardia lamblia/pathogenicity , Host-Parasite Interactions/drug effects , Host-Parasite Interactions/immunology , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Inflammasomes/drug effects , Inflammasomes/immunology , Interleukin-18/genetics , Intracellular Signaling Peptides and Proteins/genetics , Isoenzymes/pharmacology , Macrophages/drug effects , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Peptidylprolyl Isomerase/pharmacology , Phosphate-Binding Proteins/genetics , Pyroptosis/drug effects , Pyroptosis/immunology , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/immunology , Trophozoites/drug effects , Trophozoites/pathogenicity , Tumor Necrosis Factor alpha-Induced Protein 3/immunology , Ubiquitination/genetics
17.
Toxins (Basel) ; 13(12)2021 12 09.
Article En | MEDLINE | ID: mdl-34941717

Pseudomonas aeruginosa is an opportunistic, Gram-negative pathogen and an important cause of hospital acquired infections, especially in immunocompromised patients. Highly virulent P. aeruginosa strains use a type III secretion system (T3SS) to inject exoenzyme effectors directly into the cytoplasm of a target host cell. P. aeruginosa strains that express the T3SS effector, ExoU, associate with adverse outcomes in critically ill patients with pneumonia, owing to the ability of ExoU to rapidly damage host cell membranes and subvert the innate immune response to infection. Herein, we review the structure, function, regulation, and virulence characteristics of the T3SS effector ExoU, a highly cytotoxic phospholipase A2 enzyme.


Bacterial Infections/immunology , Bacterial Proteins/immunology , Host-Parasite Interactions/immunology , Immunity, Innate/drug effects , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/pathogenicity , Type III Secretion Systems/immunology , Humans
18.
Front Immunol ; 12: 770246, 2021.
Article En | MEDLINE | ID: mdl-34970259

Protozoan parasite infection causes severe diseases in humans and animals, leading to tremendous economic and medical pressure. Natural immunity is the first line of defence against parasitic infection. Currently, the role of natural host immunity in combatting parasitic infection is unclear, so further research on natural host immunity against parasites will provide a theoretical basis for the prevention and treatment of related parasitic diseases. Extracellular traps (ETs) are an important natural mechanism of immunity involving resistance to pathogens. When immune cells such as neutrophils and macrophages are stimulated by external pathogens, they release a fibrous network structure, consisting mainly of DNA and protein, that can capture and kill a variety of extracellular pathogenic microorganisms. In this review, we discuss the relevant recently reported data on ET formation induced by protozoan parasite infection, including the molecular mechanisms involved, and discuss the role of ETs in the occurrence and development of parasitic diseases.


Extracellular Traps/immunology , Immunity, Innate/immunology , Neutrophils/immunology , Protozoan Infections, Animal/immunology , Protozoan Infections/immunology , Signal Transduction/immunology , Animals , Extracellular Traps/parasitology , Host-Parasite Interactions/immunology , Humans , Leishmania/immunology , Leishmania/physiology , Neutrophils/parasitology , Plasmodium/immunology , Plasmodium/physiology , Protozoan Infections/parasitology , Protozoan Infections, Animal/parasitology , Toxoplasma/immunology , Toxoplasma/physiology
19.
PLoS Comput Biol ; 17(12): e1009714, 2021 12.
Article En | MEDLINE | ID: mdl-34932551

Hosts diverge widely in how, and how well, they defend themselves against infection and immunopathology. Why are hosts so heterogeneous? Both epidemiology and life history are commonly hypothesized to influence host immune strategy, but the relationship between immune strategy and each factor has commonly been investigated in isolation. Here, we show that interactions between life history and epidemiology are crucial for determining optimal immune specificity and sensitivity. We propose a demographically-structured population dynamics model, in which we explore sensitivity and specificity of immune responses when epidemiological risks vary with age. We find that variation in life history traits associated with both reproduction and longevity alters optimal immune strategies-but the magnitude and sometimes even direction of these effects depends on how epidemiological risks vary across life. An especially compelling example that explains previously-puzzling empirical observations is that depending on whether infection risk declines or rises at reproductive maturity, later reproductive maturity can select for either greater or lower immune specificity, potentially illustrating why studies of lifespan and immune variation across taxa have been inconclusive. Thus, the sign of selection on the life history-immune specificity relationship can be reversed in different epidemiological contexts. Drawing on published life history data from a variety of chordate taxa, we generate testable predictions for this facet of the optimal immune strategy. Our results shed light on the causes of the heterogeneity found in immune defenses both within and among species and the ultimate variability of the relationship between life history and immune specificity.


Host-Parasite Interactions/immunology , Models, Biological , Parasites , Parasitic Diseases , Animals , Biological Evolution , Humans , Longevity/immunology , Parasites/immunology , Parasites/pathogenicity , Parasitic Diseases/epidemiology , Parasitic Diseases/immunology , Parasitic Diseases/parasitology , Population Dynamics , Reproduction
20.
J Immunol Res ; 2021: 6234836, 2021.
Article En | MEDLINE | ID: mdl-34869783

Immunomodulation of airway hyperreactivity by excretory-secretory (ES) products of the first larval stage (L1) of the gastrointestinal nematode Trichuris suis is reported by us and others. Here, we aimed to identify the proteins accounting for the modulatory effects of the T. suis L1 ES proteins and studied six selected T. suis L1 proteins for their immunomodulatory efficacy in a murine OVA-induced allergic airway disease model. In particular, an enzymatically active T. suis chitinase mediated amelioration of clinical signs of airway hyperreactivity, primarily associated with suppression of eosinophil recruitment into the lung, the associated chemokines, and increased numbers of RELMα + interstitial lung macrophages. While there is no indication of T. suis chitinase directly interfering with dendritic cell activation or antigen presentation to CD4 T cells, treatment of allergic mice with the worm chitinase influenced the hosts' own chitinase activity in the inflamed lung. The three-dimensional structure of the T. suis chitinase as determined by high-resolution X-ray crystallography revealed high similarities to mouse acidic mammalian chitinase (AMCase) but a unique ability of T. suis chitinase to form dimers. Our data indicate that the structural similarities between the parasite and host chitinase contribute to the disease-ameliorating effect of the helminth-derived chitinase on allergic lung inflammation.


Chitinases/ultrastructure , Eosinophilia/drug therapy , Helminth Proteins/administration & dosage , Immunomodulating Agents/administration & dosage , Respiratory Hypersensitivity/drug therapy , Animals , Bronchoalveolar Lavage Fluid , Crystallography, X-Ray , Disease Models, Animal , Eosinophilia/diagnosis , Eosinophilia/immunology , Eosinophilia/pathology , Female , Helminth Proteins/ultrastructure , Host-Parasite Interactions/immunology , Humans , Lung/drug effects , Lung/immunology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Mice , Ovalbumin/administration & dosage , Ovalbumin/immunology , Respiratory Hypersensitivity/diagnosis , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , Trichuris/enzymology
...