Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.592
1.
Front Immunol ; 15: 1372927, 2024.
Article En | MEDLINE | ID: mdl-38742105

The parasitic helminth Schistosoma mansoni is a potent inducer of type 2 immune responses by stimulating dendritic cells (DCs) to prime T helper 2 (Th2) responses. We previously found that S. mansoni soluble egg antigens (SEA) promote the synthesis of Prostaglandin E2 (PGE2) by DCs through ERK-dependent signaling via Dectin-1 and Dectin-2 that subsequently induces OX40L expression, licensing them for Th2 priming, yet the ligands present in SEA involved in driving this response and whether specific targeting of PGE2 synthesis by DCs could affect Th2 polarization are unknown. We here show that the ability of SEA to bind Dectin-2 and drive ERK phosphorylation, PGE2 synthesis, OX40L expression, and Th2 polarization is impaired upon cleavage of high-mannose glycans by Endoglycosidase H treatment. This identifies high-mannose glycans present on glycoproteins in SEA as important drivers of this signaling axis. Moreover, we find that OX40L expression and Th2 induction are abrogated when microsomal prostaglandin E synthase-1 (mPGES) is selectively inhibited, but not when a general COX-1/2 inhibitor is used. This shows that the de novo synthesis of PGE2 is vital for the Th2 priming function of SEA-stimulated DCs as well as points to the potential existence of other COX-dependent lipid mediators that antagonize PGE2-driven Th2 polarization. Lastly, specific PGE2 inhibition following immunization with S. mansoni eggs dampened the egg-specific Th cell response. In summary, our findings provide new insights in the molecular mechanisms underpinning Th2 induction by S. mansoni and identify druggable targets for potential control of helminth driven-Th2 responses.


Antigens, Helminth , Dendritic Cells , Dinoprostone , Lectins, C-Type , Mannose , Polysaccharides , Schistosoma mansoni , Th2 Cells , Animals , Schistosoma mansoni/immunology , Dinoprostone/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Lectins, C-Type/metabolism , Lectins, C-Type/immunology , Mannose/metabolism , Mannose/immunology , Mice , Polysaccharides/immunology , Polysaccharides/metabolism , Antigens, Helminth/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/metabolism , Schistosomiasis mansoni/parasitology , Ovum/immunology , Ovum/metabolism , Mice, Inbred C57BL , OX40 Ligand/metabolism
2.
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
3.
Anal Chem ; 96(16): 6311-6320, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38594017

Schistosomiasis is a neglected tropical disease caused by worm parasites of the genus Schistosoma. Upon infection, parasite eggs can lodge inside of host organs like the liver. This leads to granuloma formation, which is the main cause of the pathology of schistosomiasis. To better understand the different levels of host-pathogen interaction and pathology, our study focused on the characterization of glycosphingolipids (GSLs). For this purpose, GSLs in livers of infected and noninfected hamsters were studied by combining high-spatial-resolution atmospheric-pressure scanning microprobe matrix-assisted laser desorption/ionization mass spectrometry imaging (AP-SMALDI MSI) with nanoscale hydrophilic interaction liquid chromatography tandem mass spectrometry (nano-HILIC MS/MS). Nano-HILIC MS/MS revealed 60 GSL species with a distinct saccharide and ceramide composition. AP-SMALDI MSI measurements were conducted in positive- and negative-ion mode for the visualization of neutral and acidic GSLs. Based on nano-HILIC MS/MS results, we discovered no downregulated but 50 significantly upregulated GSLs in liver samples of infected hamsters. AP-SMALDI MSI showed that 44 of these GSL species were associated with the granulomas in the liver tissue. Our findings suggest an important role of GSLs during granuloma formation.


Glycosphingolipids , Liver , Schistosoma mansoni , Schistosomiasis mansoni , Animals , Glycosphingolipids/metabolism , Glycosphingolipids/chemistry , Liver/metabolism , Liver/parasitology , Cricetinae , Schistosomiasis mansoni/parasitology , Schistosomiasis mansoni/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Mesocricetus , Chromatography, Liquid , Male
4.
Acta Trop ; 254: 107208, 2024 Jun.
Article En | MEDLINE | ID: mdl-38621620

The study aimed to elicit protective immune responses against murine schistosomiasis mansoni at the parasite lung- and liver stage. Two peptides showing amino acid sequence similarity to gut cysteine peptidases, which induce strong memory immune effectors in the liver, were combined with a peptide based on S. mansoni thioredoxin peroxidase (TPX), a prominent lung-stage schistosomula excretory-secretory product, and alum as adjuvant. Only one of the 2 cysteine peptidases-based peptides in a multiple antigenic peptide construct (MAP-3 and MAP-4) appeared to adjuvant protective immune responses induced by the TPX peptide in a MAP form. Production of TPX MAP-specific IgG1 serum antibodies, and increase in lung interleukin-1 (IL-1), uric acid, and reactive oxygen species (ROS) content were associated with significant (P < 0.05) 50 % reduction in recovery of lung-stage larvae. Increase in lung triglycerides and cholesterol levels appeared to provide the surviving worms with nutrients necessary for a stout double lipid bilayer barrier at the parasite-host interface. Surviving worms-released products elicited memory responses to the MAP-3 immunogen, including production of specific IgG1 antibodies and increase in liver IL-33 and ROS. Reduction in challenge worm burden recorded 45 days post infection did not exceed 48 % associated with no differences in parasite egg counts in the host liver and small intestine compared to unimmunized adjuvant control mice. Alum adjuvant assisted the second peptide, MAP-4, in production of IgG1, IgG2a, IgG2b and IgA specific antibodies and increase in liver ROS, but with no protective potential, raising doubt about the necessity of adjuvant addition. Accordingly, different vaccine formulas containing TPX MAP and 1, 2 or 3 cysteine peptidases-derived peptides with or without alum were used to immunize parallel groups of mice. Compared to unimmunized control mice, significant (P < 0.05 to < 0.005) 22 to 54 % reduction in worm burden was recorded in the different groups associated with insignificant changes in parasite egg output. The results together indicated that a schistosomiasis vaccine able to entirely prevent disease and halt its transmission still remains elusive.


Adjuvants, Immunologic , Antibodies, Helminth , Immunoglobulin G , Liver , Lung , Schistosoma mansoni , Schistosomiasis mansoni , Vaccines, Subunit , Animals , Schistosoma mansoni/immunology , Schistosomiasis mansoni/prevention & control , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Lung/parasitology , Lung/immunology , Mice , Antibodies, Helminth/immunology , Antibodies, Helminth/blood , Liver/parasitology , Liver/immunology , Immunoglobulin G/blood , Adjuvants, Immunologic/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Female , Antigens, Helminth/immunology , Disease Models, Animal , Alum Compounds/administration & dosage , Mice, Inbred BALB C , Protein Subunit Vaccines
5.
Sci Rep ; 14(1): 5974, 2024 03 12.
Article En | MEDLINE | ID: mdl-38472267

Schistosomiasis is a major Neglected Tropical Disease, caused by the infection with blood flukes in the genus Schistosoma. To complete the life cycle, the parasite undergoes asexual and sexual reproduction within an intermediate snail host and a definitive mammalian host, respectively. The intra-molluscan phase provides a critical amplification step that ensures a successful transmission. However, the cellular and molecular mechanisms underlying the development of the intra-molluscan stages remain poorly understood. Here, single cell suspensions from S. mansoni mother sporocysts were produced and sequenced using the droplet-based 10X Genomics Chromium platform. Six cell clusters comprising two tegument, muscle, neuron, parenchyma and stem/germinal cell clusters were identified and validated by in situ hybridisation. Gene Ontology term analysis predicted key biological processes for each of the clusters, including three stem/germinal sub-clusters. Furthermore, putative transcription factors predicted for stem/germinal and tegument clusters may play key roles during parasite development and interaction with the intermediate host.


Parasites , Schistosomiasis mansoni , Schistosomiasis , Animals , Gene Expression Profiling , Mammals/genetics , Mollusca/genetics , Parasites/genetics , Schistosoma mansoni/genetics , Schistosomiasis/parasitology , Schistosomiasis mansoni/parasitology
6.
J Parasitol ; 110(2): 96-105, 2024 03 01.
Article En | MEDLINE | ID: mdl-38466806

Schistosomiasis is a globally burdensome parasitic disease caused by flatworms (blood flukes) in the genus Schistosoma. The current standard treatment for schistosomiasis is the drug praziquantel, but there is an urgent need to advance novel interventions such as vaccines. Several glycolytic enzymes have been evaluated as vaccine targets for schistosomiasis, and data from these studies are reviewed here. Although these parasites are canonically considered to be intracellular, proteomic analysis has revealed that many schistosome glycolytic enzymes are additionally found at the host-interactive surface. We have recently found that the intravascular stage of Schistosoma mansoni (Sm) expresses the glycolytic enzyme phosphoglycerate mutase (PGM) on the tegumental surface. Live parasites display PGM activity, and suppression of PGM gene expression by RNA interference diminishes surface enzyme activity. Recombinant SmPGM (rSmPGM) can cleave its glycolytic substrate, 3-phosphoglycerate and can both bind to plasminogen and promote its conversion to an active form (plasmin) in vitro, suggesting a moonlighting role for this enzyme in regulating thrombosis in vivo. We found that antibodies in sera from chronically infected mice recognize rSmPGM. We also tested the protective efficacy of rSmPGM as a vaccine in the murine model. Although immunization generates high titers of anti-SmPGM antibodies (against both recombinant and native SmPGM), no significant differences in worm numbers were found between vaccinated and control animals.


Schistosomiasis mansoni , Schistosomiasis , Vaccines , Animals , Mice , Schistosoma mansoni , Phosphoglycerate Mutase , Schistosomiasis mansoni/prevention & control , Schistosomiasis mansoni/parasitology , Proteomics , Schistosomiasis/prevention & control , Antigens, Helminth , Antibodies, Helminth
7.
PLoS Negl Trop Dis ; 18(2): e0011992, 2024 Feb.
Article En | MEDLINE | ID: mdl-38416775

Schistosomiasis is a major neglected parasitic disease that affects more than 240 million people worldwide caused by Platyhelminthes of the genus Schistosoma. The treatment of schistosomiasis relies on the long-term application of a single safe drug, praziquantel (PZQ). Unfortunately, PZQ is very effective on adult parasites and poorly on larval stage and immature juvenile worms; this can partially explain the re-infection in endemic areas where patients are likely to host parasites at different developmental stages concurrently. Moreover, the risk of development of drug resistance because of the widespread use of a single drug in a large population is nowadays a serious threat. Hence, research aimed at identifying novel drugs to be used alone or in combination with PZQ is needed. Schistosomes display morphologically distinct stages during their life cycle and epigenetic mechanisms are known to play important roles in parasite growth, survival, and development. Histone deacetylase (HDAC) enzymes, particularly HDAC8, are considered valuable for therapeutic intervention for the treatment of schistosomiasis. Herein, we report the phenotypic screening on both larvae and adult Schistosoma mansoni stages of structurally different HDAC inhibitors selected from the in-house Siena library. All molecules have previously shown inhibition profiles on human HDAC6 and/or HDAC8 enzymes. Among them we identified a quinolone-based HDAC inhibitor, NF2839, that impacts larval and adult parasites as well as egg viability and maturation in vitro. Importantly, this quinolone-based compound also increases histone and tubulin acetylation in S. mansoni parasites, thus representing a leading candidate for the development of new generation anti-Schistosoma chemotherapeutics.


Anthelmintics , Histone Deacetylase Inhibitors , Quinolones , Schistosomiasis mansoni , Schistosomiasis , Animals , Humans , Anthelmintics/pharmacology , Anthelmintics/therapeutic use , Histone Deacetylase 6/antagonists & inhibitors , Larva , Praziquantel/pharmacology , Praziquantel/therapeutic use , Quinolones/pharmacology , Repressor Proteins , Schistosoma mansoni , Schistosomiasis/drug therapy , Schistosomiasis mansoni/drug therapy , Schistosomiasis mansoni/parasitology , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use
8.
Acta Trop ; 252: 107141, 2024 Apr.
Article En | MEDLINE | ID: mdl-38342286

The impact of diet composition and energy content on schistosomiasis evolution and treatment efficacy is still controversial. This study compared the impact of sucrose-rich diet and intermittent fasting on Schistosoma mansoni infection and praziquantel (PZQ)-based chemotherapy response in mice. BALB/c mice were infected with S. mansoni and followed for 15 weeks. The animals were randomized into nine groups receiving high glycemic load (high-sucrose diet - HSD), low caloric load (standard chow alternate-day fasting - ADF), and standard chow ad libitum (AL). Eight weeks after S. mansoni infection, these groups remained untreated or were treated with PZQ (300 mg/kg/day) for 3 days. Our results indicated that parasite load (S. mansoni eggs and parasite DNA levels), granulomatous inflammation (granulomas number and size), and liver microstructural damage (reduction in hepatocytes number, increase in nucleus-cytoplasm ratio, connective stroma expansion and fibrosis) were increased in ADF-treated animals. These animals also showed decreased eggs retention, granulomatous inflammation and collagen accumulation in the small intestine. Conversely, HSD diet and PZQ treatment attenuated all these parameters and stimulated hepatic regenerative response. PZQ also stimulated fibrosis resolution in HSD-treated mice, effect that was limited ADF-exposed mice. Our findings indicate that dietary glycemic and energy load can modulate schistosomiasis progression and the severity of hepatic and intestinal granulomatous inflammation in untreated and PZQ-treated mice. Thus, lower intestinal eggs retention may potentially be linked to worsening liver disease in ADF, while attenuation of hepatic and intestinal granulomatous inflammation is consistent with reduced parasite load in HSD- and PZQ-treated animals.


Anthelmintics , Liver Diseases , Schistosomiasis mansoni , Schistosomiasis , Animals , Mice , Schistosoma mansoni , Antiparasitic Agents/therapeutic use , Praziquantel/pharmacology , Schistosomiasis mansoni/drug therapy , Schistosomiasis mansoni/parasitology , Liver/parasitology , Schistosomiasis/drug therapy , Inflammation/drug therapy , Fibrosis , Diet , Sucrose/pharmacology , Sucrose/therapeutic use , Anthelmintics/therapeutic use
9.
Sci Rep ; 14(1): 1582, 2024 01 18.
Article En | MEDLINE | ID: mdl-38238498

Schistosomiasis is caused by parasites of the genus Schistosoma, which infect more than 200 million people. Praziquantel (PZQ) has been the main drug for controlling schistosomiasis for over four decades, but despite that it is ineffective against juvenile worms and size and taste issues with its pharmaceutical forms impose challenges for treating school-aged children. It is also important to note that PZQ resistant strains can be generated in laboratory conditions and observed in the field, hence its extensive use in mass drug administration programs raises concerns about resistance, highlighting the need to search for new schistosomicidal drugs. Schistosomes survival relies on the redox enzyme thioredoxin glutathione reductase (TGR), a validated target for the development of new anti-schistosomal drugs. Here we report a high-throughput fragment screening campaign of 768 compounds against S. mansoni TGR (SmTGR) using X-ray crystallography. We observed 49 binding events involving 35 distinct molecular fragments which were found to be distributed across 16 binding sites. Most sites are described for the first time within SmTGR, a noteworthy exception being the "doorstop pocket" near the NADPH binding site. We have compared results from hotspots and pocket druggability analysis of SmTGR with the experimental binding sites found in this work, with our results indicating only limited coincidence between experimental and computational results. Finally, we discuss that binding sites at the doorstop/NADPH binding site and in the SmTGR dimer interface, should be prioritized for developing SmTGR inhibitors as new antischistosomal drugs.


Multienzyme Complexes , NADH, NADPH Oxidoreductases , Schistosomiasis mansoni , Schistosomiasis , Animals , Child , Humans , Schistosoma mansoni , Crystallography, X-Ray , NADP/metabolism , Schistosomiasis/drug therapy , Binding Sites , Schistosomiasis mansoni/parasitology
10.
J Helminthol ; 98: e3, 2024 Jan 03.
Article En | MEDLINE | ID: mdl-38167243

Schistosomiasis afflicts approximately 120 million individuals globally. The hepatic pathology that occurs due to egg-induced granuloma and fibrosis is commonly attributed to this condition. However, there is currently no efficacious treatment available for either of these conditions.Our study aimed to investigate the potential antifibrotic and antiparasitic properties of different doses of gallic acid (GA) in experimental schistosomiasis mansoni. In addition, we investigated the outcomes of co-administering it with the standard anti-schistosomiasis treatment, praziquantel (PZQ).In experiment I, Schistosoma mansoni-infected mice were administered GA at doses of 10, 20, or 40 mg/kg. Their effectiveness was evaluated through parasitological (worm and egg loads, granuloma number and diameter), pathological (fibrosis percentage and H-score of hepatic stellate cells (HSCs)), and functional (liver enzymes) tests. In experiment II, we investigated the optimal dosage that yielded the best outcomes. This dosage was administered in conjunction with PZQ and was evaluated regarding the parasitological, pathological, functional, and immunological (fibrosis-regulating cytokines) activities.Our findings indicate that the administration of 40 mg/kg GA exhibited the highest level of effectiveness in experiment I. In experiment II, it exhibited lower antiparasitic efficacy in comparison to PZQ. However, it surpassed PZQ in other tests. It showed enhanced outcomes when combined with PZQ.In conclusion, our findings reveal that GA only slightly increased the antischistosomal activity of PZQ. However, it was linked to decreased fibrosis, particularly when administrated with PZQ. Our pilot study identifies GA as a natural antifibrotic agent, which could be administered with PZQ to mitigate the development of fibrosis.


Anthelmintics , Schistosomiasis mansoni , Animals , Mice , Schistosomiasis mansoni/parasitology , Antiparasitic Agents/pharmacology , Antiparasitic Agents/therapeutic use , Gallic Acid/pharmacology , Gallic Acid/therapeutic use , Pilot Projects , Liver/parasitology , Praziquantel , Schistosoma mansoni , Fibrosis , Granuloma/drug therapy , Granuloma/pathology
11.
Parasitol Int ; 99: 102833, 2024 Apr.
Article En | MEDLINE | ID: mdl-38061487

The WHO considers schistosomiasis, which is controlled by the mass administration of the drug praziquantel (PZQ), to be a neglected tropical disease. Despite its clinical use for over four decades, PZQ remains the only choice of chemotherapy against this disease. Regarding the previous studies that demonstrated that PZQ activates the transient receptor potential (TRP) channel in Schistosoma mansoni (Sm.TRPMPZQ), the expression profile of the ortholog of this channel gene (Smp_246790.5) in S. japonicum (EWB00_008853) (Sj.TRPMPZQ) was analyzed. The relative expression of this gene in various stages of the parasite lifecycle was analyzed by quantitative real-time reverse transcription-PCR (qRT-PCR), and the expression of Sj.TRPMPZQ was observed by immunohistochemical staining using anti-serum against the recombinant Sj.TRPMPZQ protein. qRT-PCR revealed the significantly lower mRNA expression in the snail stage in comparison to other stages (p < 0.01). The relative quantity of the Sj.TRPMPZQ expression for paired females, unpaired males, and eggs was 60%, 56%, and 68%, respectively, in comparison to paired males that showed the highest expression (p < 0.05). Interestingly, immunostaining demonstrated that Sj.TRPMPZQ is expressed in the parenchyma which contains muscle cells, neuronal cells and tegument cells in adult worms. This may support the two major effects of PZQ-worm paralysis and tegument disruption-induced by channel activation. Moreover, the channel was expressed in both the eggshell and the miracidia inside, but could not be observed in sporocyst. These results suggest that the expression of Sj.TRPMPQZ corresponds to the known sensitivity of S. japonicum to PZQ.


Anthelmintics , Schistosoma japonicum , Schistosomiasis japonica , Schistosomiasis mansoni , TRPM Cation Channels , Male , Female , Animals , Praziquantel , Schistosoma japonicum/physiology , Schistosoma mansoni/genetics , Schistosomiasis japonica/parasitology , Schistosomiasis mansoni/parasitology , Anthelmintics/pharmacology , Anthelmintics/therapeutic use
12.
Microbiol Spectr ; 12(1): e0219323, 2024 Jan 11.
Article En | MEDLINE | ID: mdl-38047698

IMPORTANCE: Schistosomes cause schistosomiasis, one of the neglected tropical diseases as defined by the WHO. For decades, the treatment of schistosomiasis relies on a single drug, praziquantel. Due to its wide use, there is justified fear of resistance against this drug, and a vaccine is not available. Besides its biological relevance in signal transduction processes, the class of G protein-coupled receptors (GPCRs) is also well suited for drug design. Against this background, we characterized one GPCR of Schistosoma mansoni, SmGPCR20, at the molecular and functional level. We identified two potential neuropeptides (NPPs) as ligands, SmNPP26 and SmNPP40, and unraveled their roles, in combination with SmGPCR20, in neuronal processes controlling egg production, oogenesis, and growth of S. mansoni females. Since eggs are closely associated with the pathogenesis of schistosomiasis, our results contribute to the understanding of processes leading to egg production in schistosomes, which is under the control of pairing in this exceptional parasite.


Schistosomiasis mansoni , Schistosomiasis , Animals , Female , Schistosoma mansoni , Schistosomiasis mansoni/parasitology , Schistosomiasis mansoni/pathology , Rhodopsin , Sex Differentiation
13.
PLoS Negl Trop Dis ; 17(12): e0011776, 2023 Dec.
Article En | MEDLINE | ID: mdl-38039267

BACKGROUND: CLA (conjugated linoleic acid)-mediated activation of the schistosome tegument-associated sphingomyelinase and consequent disruption of the outer membrane might allow host antibodies to access the apical membrane antigens. Here, we investigated a novel approach to enhance specific antibody delivery to concealed surface membrane antigens of Schistosoma mansoni utilising antibody-conjugated-CLA nanomicelle technology. METHODOLOGY/PRINCIPAL FINDINGS: We invented and characterised an amphiphilic CLA-loaded whey protein co-polymer (CLA-W) as an IV injectable protein nanocarrier. Rabbit anti-Schistosoma mansoni infection (anti-SmI) and anti-Schistosoma mansoni alkaline phosphatase specific IgG antibodies were purified from rabbit sera and conjugated to the surface of CLA-W co-polymer to form antibody-conjugated-CLA-W nanomicelles (Ab-CLA-W). We investigated the schistosomicidal effects of CLA-W and Ab-CLA-W in a mouse model of Schistosoma mansoni against early and late stages of infection. Results showed that conjugation of nanomicelles with antibodies, namely anti-SmI, significantly enhanced the micelles' schistosomicidal and anti-pathology activities at both the schistosomula and adult worm stages of the infection resulting in 64.6%-89.9% reductions in worm number; 72.5-94% and 66.4-85.2% reductions in hepatic eggs and granulomas, respectively. Treatment induced overall improvement in liver histopathology, reducing granuloma size and fibrosis and significantly affecting egg viability. Indirect immunofluorescence confirmed CLA-W-mediated antigen exposure on the worm surface. Electron microscopy revealed extensive ultrastructural damage in worm tegument induced by anti-SmI-CLA-W. CONCLUSION/SIGNIFICANCE: The novel antibody-targeted nano-sized CLA delivery system offers great promise for treatment of Schistosoma mansoni infection and control of its transmission. Our in vivo observations confirm an immune-mediated enhanced effect of the schistosomicidal action of CLA and hints at the prospect of nanotechnology-based immunotherapy, not only for schistosomiasis, but also for other parasitic infections in which chemotherapy has been shown to be immune-dependent. The results propose that the immunodominant reactivity of the anti-SmI serum, Schistosoma mansoni fructose biphosphate aldolase, SmFBPA, merits serious attention as a therapeutic and vaccine candidate.


Schistosomiasis mansoni , Schistosomiasis , Schistosomicides , Mice , Animals , Rabbits , Schistosomiasis mansoni/parasitology , Schistosoma mansoni , Schistosomiasis/drug therapy , Antibodies, Helminth , Schistosomicides/pharmacology , Polymers/pharmacology , Polymers/therapeutic use , Antigens, Helminth
14.
Parasitol Res ; 123(1): 72, 2023 Dec 27.
Article En | MEDLINE | ID: mdl-38148420

Probiotics contribute to the integrity of the intestinal mucosa and preventing dysbiosis caused by opportunistic pathogens, such as intestinal helminths. Bacillus cereus GM obtained from Biovicerin® was cultured to obtain spores for in vivo evaluation on experimental schistosomiasis. The assay was performed for 90 days, where all animals were infected with 50 cercariae of Schistosoma mansoni on the 15th day. Three experimental groups were formed, as follows: G1-saline solution from the 1st until the 90th day; G2-B. cereus GM (105 spores in 300 µL of sterile saline) from the 1st until the 90th day; and G3-B. cereus GM 35th day (onset of oviposition) until the 90th day. G2 showed a significant reduction of 43.4% of total worms, 48.8% of female worms and 42.5% of eggs in the liver tissue. In G3, the reduction was 25.2%, 29.1%, and 44% of the total number of worms, female worms, and eggs in the liver tissue, respectively. G2 and G3 showed a 25% (p < 0.001) and 22% (p < 0.001) reduction in AST levels, respectively, but ALT levels did not change. ALP levels were reduced by 23% (p < 0.001) in the G2 group, but not in the G3. The average volume of granulomas reduced (p < 0.0001) 65.2% and 46.3% in the liver tissue and 83.0% and 53.2% in the intestine, respectively, in groups G2 and G3. Th1 profile cytokine (IFN-γ, TNF-α, and IL-6) and IL-17 were significantly increased (p < 0.001) stimulated with B. cereus GM in groups G2 and G3. IL-4 showed significant values when the stimulus was mediated by ConA. By modulating the immune response, B. cereus GM reduced the burden of worms, improved some markers of liver function, and reduced the granulomatous inflammatory reaction in mice infected with S. mansoni, especially when administered before infection.


Probiotics , Schistosomiasis mansoni , Schistosomiasis , Female , Animals , Mice , Schistosomiasis mansoni/parasitology , Bacillus cereus , Schistosoma mansoni , Schistosomiasis/parasitology , Liver/parasitology
15.
Sci Rep ; 13(1): 20390, 2023 11 21.
Article En | MEDLINE | ID: mdl-37990129

Schistosomiasis is a parasitic disease affecting more than 250 million people worldwide. The transcription factor c-Jun, which is induced in S. mansoni infection-associated liver disease, can promote hepatocyte survival but can also trigger hepatocellular carcinogenesis. We aimed to analyze the hepatic role of c-Jun following S. mansoni infection. We adopted a hepatocyte-specific c-Jun knockout mouse model (Alb-Cre/c-Jun loxP) and analyzed liver tissue and serum samples by quantitative real-time PCR array, western blotting, immunohistochemistry, hydroxyproline quantification, and functional analyses. Hepatocyte-specific c-Jun knockout (c-JunΔli) was confirmed by immunohistochemistry and western blotting. Infection with S. mansoni induced elevated aminotransferase-serum levels in c-JunΔli mice. Of note, hepatic Cyclin D1 expression was induced in infected c-Junf/f control mice but to a lower extent in c-JunΔli mice. S. mansoni soluble egg antigen-induced proliferation in a human hepatoma cell line was diminished by inhibition of c-Jun signaling. Markers for apoptosis, oxidative stress, ER stress, inflammation, autophagy, DNA-damage, and fibrosis were not altered in S. mansoni infected c-JunΔli mice compared to infected c-Junf/f controls. Enhanced liver damage in c-JunΔli mice suggested a protective role of c-Jun. A reduced Cyclin D1 expression and reduced hepatic regeneration could be the reason. In addition, it seems likely that the trends in pathological changes in c-JunΔli mice cumulatively led to a loss of the protective potential being responsible for the increased hepatocyte damage and loss of regenerative ability.


Schistosoma mansoni , Schistosomiasis mansoni , Humans , Mice , Animals , Cyclin D1/metabolism , Schistosomiasis mansoni/parasitology , Liver/metabolism , Hepatocytes/metabolism , Cell Proliferation
16.
Sci Data ; 10(1): 775, 2023 11 07.
Article En | MEDLINE | ID: mdl-37935722

The flatworm Schistosoma mansoni is an important but neglected pathogen that causes the disease schistosomiasis in millions of people worldwide. The parasite has a complex life cycle, undergoing sexual reproduction in a mammalian host and asexual replication in a snail host. Understanding the molecular mechanisms that the parasite uses to transition between hosts and develop into dimorphic reproductively competent adults may reveal new strategies for control. We present the first comprehensive transcriptomic analysis of S. mansoni, from eggs to sexually naïve worms. Focusing on eight life stages spanning free-living water-borne and parasitic stages from both intermediate and definitive hosts, we have generated deep RNA-seq data for five replicates per group for a total of 75 data sets. The data were produced using a single approach to increase the accuracy of stage-to-stage comparisons and made accessible via a user-friendly tool to visualise and explore gene expression ( https://lifecycle.schisto.xyz/ ). These data are valuable for understanding the biology and sex-specific development of schistosomes and the interpretation of complementary genomic and functional genetics studies.


Schistosoma mansoni , Transcriptome , Animals , Female , Humans , Male , Gene Expression Profiling , Schistosoma mansoni/genetics , Schistosomiasis mansoni/metabolism , Schistosomiasis mansoni/parasitology , Sex Factors
17.
Am J Trop Med Hyg ; 109(3): 667-675, 2023 09 06.
Article En | MEDLINE | ID: mdl-37580029

Soil-transmitted helminths (STHs) and Schistosoma mansoni infections are common parasitic infections in sub-Saharan Africa. Their distributions vary across altitudes in Ethiopia. Hence, ongoing updates of infection risk factors and prevalence data are necessary for successful intervention. A school-based, cross-sectional study was conducted from October to December 2019 involving 530 schoolchildren who were recruited by systematic random sampling from Amhara Regional State. A structured questionnaire was used to collect data on sociodemographic, geographical and environmental factors. Stool samples were collected and processed by Ritchie's and Kato Katz techniques. Data were entered into EpiData and analyzed using SPSS. Descriptive statistics were used to compute prevalence, and logistic regression was used to assess factors associated with STHs and S. mansoni infections. Variables with P < 0.05 were considered statistically significant. Among 530 schoolchildren, 169 (31.9%) and 78 (14.7%) were infected with STHs and S. mansoni, respectively. The prevalence of STHs (40.2%) in the semi-highlands and S. mansoni (30.4%) in the lowlands was high. Infrequent shoes wearing, exposure to soil, not washing vegetables before eating, and living outside in lowland areas were significantly associated with STHs infections (P < 0.05). Schoolchildren who fetched water to irrigate fields, swam in rivers, and lived in lowland areas were significantly associated with S. mansoni infection (P < 0.05). In conclusion, prevalence rates of STHs and S. mansoni infections differed across altitudes in Amhara Regional State. Therefore, current control strategies including deworming and provision of clean water and education on sanitation and hygiene should be intensified and adapted to the local context.


Helminthiasis , Helminths , Schistosomiasis mansoni , Animals , Humans , Child , Schistosoma mansoni , Ethiopia/epidemiology , Soil/parasitology , Prevalence , Cross-Sectional Studies , Schistosomiasis mansoni/epidemiology , Schistosomiasis mansoni/parasitology , Helminthiasis/epidemiology , Helminthiasis/parasitology , Water , Feces/parasitology
18.
Microbiol Spectr ; 11(4): e0139323, 2023 08 17.
Article En | MEDLINE | ID: mdl-37409934

Schistosomiasis is a parasitic disease that afflicts approximately 250 million people worldwide. There is an urgent demand for new antiparasitic agents because praziquantel, the only drug available for the treatment of schistosomiasis, is not universally effective and may derail current progress toward the WHO goal of eliminating this disease as a public health problem by 2030. Nifuroxazide (NFZ), an oral nitrofuran antibiotic, has recently been explored to be repurposed for parasitic diseases. Here, in vitro, in vivo, and in silico studies were conducted to evaluate the activity of NFZ on Schistosoma mansoni. The in vitro study showed significant antiparasitic activity, with 50% effective concentration (EC50) and 90% effective concentration (EC90) values of 8.2 to 10.8 and 13.7 to 19.3 µM, respectively. NFZ also affected worm pairing and egg production and induced severe damage to the tegument of schistosomes. In vivo, a single oral dose of NFZ (400 mg/kg of body weight) to mice harboring either prepatent or patent S. mansoni infection significantly reduced the total worm burden (~40%). In patent infection, NFZ achieved a high reduction in the number of eggs (~80%), but the drug caused a low reduction in the egg burden of animals with prepatent infection. Finally, results from in silico target fishing methods predicted that serine/threonine kinases could be one of the potential targets for NFZ in S. mansoni. Overall, the present study revealed that NFZ possesses antischistosomal properties, mainly in terms of egg burden reduction in animals with patent S. mansoni infection. IMPORTANCE The increasing recognition of the burden imposed by helminthiasis, associated with the limited therapeutic arsenal, has led to initiatives and strategies to research and develop new drugs for the treatment of schistosomiasis. One of these strategies is drug repurposing, which considers low-risk compounds with potentially reduced costs and shorter time for development. In this study, nifuroxazide (NFZ) was evaluated for its anti-Schistosoma mansoni potential through in vitro, in vivo, and in silico studies. In vitro, NFZ affected worm pairing and egg production and induced severe damage to the tegument of schistosomes. In vivo, a single oral dose of NFZ (400 mg/kg) to mice harboring either prepatent or patent S. mansoni infection significantly reduced the total worm burden and egg production. In silico investigations have identified serine/threonine kinases as a molecular target for NFZ. Collectively, these results implied that NFZ might be a potential therapeutic candidate for the treatment of schistosomiasis.


Nitrofurans , Schistosomiasis mansoni , Schistosomiasis , Schistosomicides , Animals , Mice , Schistosomicides/pharmacology , Schistosomicides/therapeutic use , Schistosomiasis mansoni/drug therapy , Schistosomiasis mansoni/parasitology , Schistosoma mansoni , Nitrofurans/pharmacology , Nitrofurans/therapeutic use , Threonine/pharmacology , Threonine/therapeutic use , Serine
19.
J Infect Dis ; 228(9): 1299-1303, 2023 11 02.
Article En | MEDLINE | ID: mdl-37487539

While symbiotic relationships between invertebrates and bacteria have been extensively described, studies of microbial communities inhabiting parasitic worms remain scarce. Exploring the microbiota associated with helminths responsible for major infectious diseases will inform on parasite biology, host-pathogen interactions, and disease pathophysiology. We investigated the presence of microorganisms inhabiting tissues of the human parasite Schistosoma mansoni. In situ hybridization using a pan-bacterial 16S rRNA gene probe revealed bacteria colonizing key developmental stages that were successfully removed after antibiotic treatment of live parasites. Understanding the composition and function of the S. mansoni-associated microbiota may lead to the development of novel microbiome-targeting control strategies.


Helminths , Parasites , Schistosomiasis mansoni , Animals , Humans , Schistosoma mansoni/genetics , Parasites/genetics , RNA, Ribosomal, 16S/genetics , Life Cycle Stages , Bacteria/genetics , Schistosomiasis mansoni/parasitology
20.
PLoS Pathog ; 19(7): e1011018, 2023 07.
Article En | MEDLINE | ID: mdl-37428793

Human schistosomiasis is a neglected tropical disease caused by Schistosoma mansoni, S. haematobium, and S. japonicum. Praziquantel (PZQ) is the method of choice for treatment. Due to constant selection pressure, there is an urgent need for new therapies for schistosomiasis. Previous treatment of S. mansoni included the use of oxamniquine (OXA), a drug that is activated by a schistosome sulfotransferase (SULT). Guided by data from X-ray crystallography and Schistosoma killing assays more than 350 OXA derivatives were designed, synthesized, and tested. We were able to identify CIDD-0150610 and CIDD-0150303 as potent derivatives in vitro that kill (100%) of all three Schistosoma species at a final concentration of 71.5 µM. We evaluated the efficacy of the best OXA derivates in an in vivo model after treatment with a single dose of 100 mg/kg by oral gavage. The highest rate of worm burden reduction was achieved by CIDD -150303 (81.8%) against S. mansoni, CIDD-0149830 (80.2%) against S. haematobium and CIDD-066790 (86.7%) against S. japonicum. We have also evaluated the ability of the derivatives to kill immature stages since PZQ does not kill immature schistosomes. CIDD-0150303 demonstrated (100%) killing for all life stages at a final concentration of 143 µM in vitro and effective reduction in worm burden in vivo against S. mansoni. To understand how OXA derivatives fit in the SULT binding pocket, X-ray crystal structures of CIDD-0150303 and CIDD-0150610 demonstrate that the SULT active site will accommodate further modifications to our most active compounds as we fine tune them to increase favorable pharmacokinetic properties. Treatment with a single dose of 100 mg/kg by oral gavage with co-dose of PZQ + CIDD-0150303 reduced the worm burden of PZQ resistant parasites in an animal model by 90.8%. Therefore, we conclude that CIDD-0150303, CIDD-0149830 and CIDD-066790 are novel drugs that overcome some of PZQ limitations, and CIDD-0150303 can be used with PZQ in combination therapy.


Anthelmintics , Schistosomiasis mansoni , Schistosomiasis , Animals , Humans , Praziquantel/pharmacology , Praziquantel/chemistry , Oxamniquine/pharmacology , Schistosomiasis/drug therapy , Schistosomiasis/parasitology , Schistosoma mansoni , Combined Modality Therapy , Neglected Diseases/drug therapy , Schistosomiasis mansoni/drug therapy , Schistosomiasis mansoni/parasitology
...