Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 866
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 300(1): 105528, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38043794

RESUMO

Parasitic flatworms cause various clinical and veterinary infections that impart a huge burden worldwide. The most clinically impactful infection is schistosomiasis, a neglected tropical disease caused by parasitic blood flukes. Schistosomiasis is treated with praziquantel (PZQ), an old drug introduced over 40 years ago. New drugs are urgently needed, as while PZQ is broadly effective it suffers from several limitations including poor efficacy against juvenile worms, which may prevent it from being completely curative. An old compound that retains efficacy against juvenile worms is the benzodiazepine meclonazepam (MCLZ). However, host side effects caused by benzodiazepines preclude development of MCLZ as a drug and MCLZ lacks an identified parasite target to catalyze rational drug design for engineering out human host activity. Here, we identify a transient receptor potential ion channel of the melastatin subfamily, named TRPMMCLZ, as a parasite target of MCLZ. MCLZ potently activates Schistosoma mansoni TRPMMCLZ through engagement of a binding pocket within the voltage-sensor-like domain of the ion channel to cause worm paralysis, tissue depolarization, and surface damage. TRPMMCLZ reproduces all known features of MCLZ action on schistosomes, including a lower activity versus Schistosoma japonicum, which is explained by a polymorphism within this voltage-sensor-like domain-binding pocket. TRPMMCLZ is distinct from the TRP channel targeted by PZQ (TRPMPZQ), with both anthelmintic chemotypes targeting unique parasite TRPM paralogs. This advances TRPMMCLZ as a novel druggable target that could circumvent any target-based resistance emerging in response to current mass drug administration campaigns centered on PZQ.


Assuntos
Anti-Helmínticos , Clonazepam , Esquistossomose mansoni , Canais de Cátion TRPM , Animais , Humanos , Anti-Helmínticos/farmacologia , Benzodiazepinas/farmacologia , Benzodiazepinonas/farmacologia , Clonazepam/análogos & derivados , Clonazepam/farmacologia , Praziquantel/farmacologia , Schistosoma mansoni/efeitos dos fármacos , Schistosoma mansoni/metabolismo , Esquistossomose mansoni/tratamento farmacológico , Canais de Cátion TRPM/agonistas
2.
BMC Infect Dis ; 24(1): 636, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38918706

RESUMO

BACKGROUND: Schistosomiasis is a parasitic disease caused by trematodes of the genus Schistosoma. The intravascular worms acquire the nutrients necessary for their survival from host blood. Since all animals are auxotrophic for riboflavin (vitamin B2), schistosomes too must import it to survive. Riboflavin is an essential component of the coenzymes flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD); these support key functions of dozens of flavoenzymes. METHODS: Here, using a combination of metabolomics, enzyme kinetics and in silico molecular analysis, we focus on the biochemistry of riboflavin and its metabolites in Schistosoma mansoni (Sm). RESULTS: We show that when schistosomes are incubated in murine plasma, levels of FAD decrease over time while levels of FMN increase. We show that live schistosomes cleave exogenous FAD to generate FMN and this ability is significantly blocked when expression of the surface nucleotide pyrophosphatase/phosphodiesterase ectoenzyme SmNPP5 is suppressed using RNAi. Recombinant SmNPP5 cleaves FAD with a Km of 178 ± 5.9 µM and Kcat/Km of 324,734 ± 36,347 M- 1.S- 1. The FAD-dependent enzyme IL-4I1 drives the oxidative deamination of phenylalanine to produce phenylpyruvate and H2O2. Since schistosomes are damaged by H2O2, we determined if SmNPP5 could impede H2O2 production by blocking IL-4I1 action in vitro. We found that this was not the case; covalently bound FAD on IL-4I1 appears inaccessible to SmNPP5. We also report that live schistosomes can cleave exogenous FMN to generate riboflavin and this ability is significantly impeded when expression of a second surface ectoenzyme (alkaline phosphatase, SmAP) is suppressed. Recombinant SmAP cleaves FMN with a Km of 3.82 ± 0.58 mM and Kcat/Km of 1393 ± 347 M- 1.S- 1. CONCLUSIONS: The sequential hydrolysis of FAD by tegumental ecto-enzymes SmNPP5 and SmAP can generate free vitamin B2 around the worms from where it can be conveniently imported by the recently described schistosome riboflavin transporter SmaRT. Finally, we identified in silico schistosome homologs of enzymes that are involved in intracellular vitamin B2 metabolism. These are riboflavin kinase (SmRFK) as well as FAD synthase (SmFADS); cDNAs encoding these two enzymes were cloned and sequenced. SmRFK is predicted to convert riboflavin to FMN while SmFADS could further act on FMN to regenerate FAD in order to facilitate robust vitamin B2-dependent metabolism in schistosomes.


Assuntos
Mononucleotídeo de Flavina , Flavina-Adenina Dinucleotídeo , Riboflavina , Schistosoma mansoni , Riboflavina/metabolismo , Mononucleotídeo de Flavina/metabolismo , Animais , Flavina-Adenina Dinucleotídeo/metabolismo , Schistosoma mansoni/metabolismo , Schistosoma mansoni/genética , Camundongos , Humanos , Esquistossomose mansoni/parasitologia , Esquistossomose mansoni/metabolismo
3.
Inflammopharmacology ; 32(1): 763-775, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38041753

RESUMO

Hepatic schistosomiasis is a prevalent form of chronic liver disease that drastically affects human health. Nevertheless, an antifibrotic drug that could suppress the development of hepatic fibrosis does not exist yet. The current study aimed to evaluate the effect of resveratrol, a natural polyphenol with multiple biological activities, on Schistosoma mansoni (S. mansoni)-induced hepatic fibrosis and delineate the underlying molecular mechanism. Swiss male albino mice were randomly assigned into infected and non-infected groups. Hepatic schistosomiasis infection was induced via exposure to S. mansoni cercariae. 6 weeks later, resveratrol was administrated either as 20 mg/kg/day or 100 mg/kg/day for 4 weeks to two infected groups. Another group received vehicle and served as infected control group. At the end of the study, portal hemodynamic, biochemical, and histopathological evaluation of liver tissues were conducted. Remarkably, resveratrol significantly reduced portal pressure, portal and mesenteric flow in a dose-dependent manner. It improved several key features of hepatic injury as evidenced biochemically by a significant reduction of bilirubin and liver enzymes, and histologically by amelioration of the granulomatous and inflammatory reactions. In line, resveratrol reduced the expression of pro-inflammatory markers; TNF-α, IL-1ß and MCP-1 mRNA, together with fibrotic markers; collagen-1, TGF-ß1 and α-SMA. Moreover, resveratrol restored SIRT1/NF-κB balance in hepatic tissues which is the main switch-off control for all the fibrotic and inflammatory mechanisms. Taken together, it can be inferred that resveratrol possesses a possible anti-fibrotic effect that can halt the progression of hepatic schistosomiasis via targeting SIRT1/ NF-κB signaling.


Assuntos
Schistosoma mansoni , Esquistossomose , Camundongos , Animais , Masculino , Humanos , Schistosoma mansoni/metabolismo , NF-kappa B/metabolismo , Resveratrol/farmacologia , Sirtuína 1 , Cirrose Hepática/tratamento farmacológico
4.
Biochemistry ; 62(9): 1497-1508, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-37071546

RESUMO

Thioredoxin/glutathione reductase from Schistosoma mansoni (SmTGR) catalyzes the reduction of both oxidized thioredoxin and glutathione with electrons from reduced nicotinamide adenine dinucleotide phosphate (NADPH). SmTGR is a drug target for the treatment of Schistosomiasis, an infection caused by Schistosoma platyhelminths residing in the blood vessels of the host. Schistosoma spp. are reliant on TGR enzymes as they lack catalase and so use reduced thioredoxin and glutathione to regenerate peroxiredoxins consumed in the detoxification of reactive oxygen species. SmTGR is a flavin adenine dinucleotide (FAD)-dependent enzyme, and we have used the flavin as a spectrophotometric reporter to observe the movement of electrons within the enzyme. The data show that NADPH fractionally reduces the active site flavin with an observed rate constant estimated in this study to be ∼3000 s-1. The flavin then reoxidizes by passing electrons at a similar rate to the proximal Cys159-Cys154 disulfide pair. The dissociation of NADP+ occurs with a rate of ∼180 s-1, which induces the deprotonation of Cys159, and this coincides with the accumulation of an intense FAD-thiolate charge transfer band. It is proposed that the electrons then pass to the Cys596-Cys597 disulfide pair of the associated subunit in the dimer with a net rate constant of ∼2 s-1. (Note: Cys597 is Sec597 in wild-type (WT) SmTGR.) From this position, the electrons can be passed to oxidized thioredoxin or further into the protein to reduce the Cys28-Cys31 disulfide pair of the originating subunit of the dimer. From the Cys28-Cys31 center, electrons can then pass to oxidized glutathione that has a binding site directly adjacent.


Assuntos
Flavina-Adenina Dinucleotídeo , Schistosoma mansoni , Animais , Schistosoma mansoni/metabolismo , Glutationa Redutase/metabolismo , NADP/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Tiorredoxina Dissulfeto Redutase/metabolismo , Glutationa/metabolismo , Dissulfetos , Tiorredoxinas/metabolismo , Oxirredução
5.
PLoS Pathog ; 17(12): e1010140, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34910770

RESUMO

Schistosomes infect over 200 million of the world's poorest people, but unfortunately treatment relies on a single drug. Nuclear hormone receptors are ligand-activated transcription factors that regulate diverse processes in metazoans, yet few have been functionally characterized in schistosomes. During a systematic analysis of nuclear receptor function, we found that an FTZ-F1-like receptor was essential for parasite survival. Using a combination of transcriptional profiling and chromatin immunoprecipitation (ChIP), we discovered that the micro-exon gene meg-8.3 is a transcriptional target of SmFTZ-F1. We found that both Smftz-f1 and meg-8.3 are required for esophageal gland maintenance as well as integrity of the worm's head. Together, these studies define a new role for micro-exon gene function in the parasite and suggest that factors associated with the esophageal gland could represent viable therapeutic targets.


Assuntos
Esôfago/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas de Helminto/metabolismo , Schistosoma mansoni/metabolismo , Fatores de Transcrição/metabolismo , Animais
6.
PLoS Pathog ; 17(1): e1009198, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33417618

RESUMO

Macrophages have a defined role in the pathogenesis of metabolic disease and cholesterol metabolism where alternative activation of macrophages is thought to be beneficial to both glucose and cholesterol metabolism during high fat diet induced disease. It is well established that helminth infection protects from metabolic disease, but the mechanisms underlying protection are not well understood. Here, we investigated the effects of Schistosoma mansoni infection and cytokine activation in the metabolic signatures of bone marrow derived macrophages using an approach that integrated transcriptomics, metabolomics, and lipidomics in a metabolic disease prone mouse model. We demonstrate that bone marrow derived macrophages (BMDM) from S. mansoni infected male ApoE-/- mice have dramatically increased mitochondrial respiration compared to those from uninfected mice. This change is associated with increased glucose and palmitate shuttling into TCA cycle intermediates, increased accumulation of free fatty acids, and decreased accumulation of cellular cholesterol esters, tri and diglycerides, and is dependent on mgll activity. Systemic injection of IL-4 complexes is unable to recapitulate either reductions in systemic glucose AUC or the re-programing of BMDM mitochondrial respiration seen in infected males. Importantly, the metabolic reprogramming of male myeloid cells is transferrable via bone marrow transplantation to an uninfected host, indicating maintenance of reprogramming in the absence of sustained antigen exposure. Finally, schistosome induced metabolic and bone marrow modulation is sex-dependent, with infection protecting male, but not female mice from glucose intolerance and obesity. Our findings identify a transferable, long-lasting sex-dependent reprograming of the metabolic signature of macrophages by helminth infection, providing key mechanistic insight into the factors regulating the beneficial roles of helminth infection in metabolic disease.


Assuntos
Antígenos/imunologia , Linhagem da Célula , Macrófagos/metabolismo , Doenças Metabólicas/prevenção & controle , Células Mieloides/metabolismo , Schistosoma mansoni/metabolismo , Esquistossomose mansoni/metabolismo , Animais , Reprogramação Celular , Dieta Hiperlipídica/efeitos adversos , Feminino , Metabolismo dos Lipídeos , Macrófagos/imunologia , Macrófagos/parasitologia , Masculino , Doenças Metabólicas/imunologia , Doenças Metabólicas/parasitologia , Metaboloma , Camundongos , Camundongos Knockout para ApoE , Células Mieloides/imunologia , Células Mieloides/parasitologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/parasitologia
7.
Rapid Commun Mass Spectrom ; 38 Suppl 1: e9523, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37070167

RESUMO

Schistosomes are blood flukes with specialised tissues and organs, each one playing a pivotal role in perpetuating the parasite life cycle. Herein, we describe a detailed methodology for preserving the proteome of adult Schistosoma mansoni worms during manual dissection for enrichment of tissues associated with the parasite's alimentary tract. We provide step-by-step directions for specimen storage and dissection while in preservative solution, tissue homogenisation, protein extraction and digestion using a methodology fully compatible with downstream quantitative liquid chromatography-mass spectrometry analysis. Our methodology uses label-free and QconCAT-based absolute quantification for detection of S. mansoni oesophageal gland products proposed as vaccine candidates. Through stabilisation of the proteome and minimising sample degradation during dissection our approach has allowed us to access the hidden proteome of target tissues not readily available from total lysates because of their small volume. This protocol can be replicated or adapted to other Schistosoma species lacking quantitative proteomics characterisation of specialised tissues for discovery of proteins with potential diagnostic and therapeutic utility.


Assuntos
Espectrometria de Massa com Cromatografia Líquida , Proteômica , Animais , Proteômica/métodos , Cromatografia Líquida , Proteoma/metabolismo , Espectrometria de Massas em Tandem , Schistosoma mansoni/química , Schistosoma mansoni/metabolismo
8.
J Invertebr Pathol ; 201: 107994, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37741506

RESUMO

Despite undergoing development within a germfree egg capsule, embryos and larvae of the freshwater snail Biomphalaria glabrata possess passive immune protection in the form of parentally-derived antimicrobial proteins in the perivitelline fluid. However, the point at which larvae begin to form their own internal defense system (IDS), which consists of both plasma proteins and hemocytes, is not known. In this study, hemocyte-like cells were observed in mechanically-disrupted late trochophores and veligers of the BS-90 strain of B. glabrata. These cells showed the properties of glass adherence, spreading, motility, and binding and phagocytosing polystyrene microspheres. No hemocyte-like cells were recovered from the early trochophore stage, and therefore their formation first occurs during subsequent maturation. Numbers of hemocyte-like cells increased during larval development. Although the functional significance of these cells is not known, they may represent the initial cellular component of the IDS.


Assuntos
Biomphalaria , Animais , Hemócitos , Schistosoma mansoni/metabolismo , Larva , Caramujos , Água Doce
9.
J Biol Chem ; 297(2): 100979, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34303703

RESUMO

Schistosomiasis, a neglected tropical disease caused by trematodes of the Schistosoma genus, affects over 250 million people around the world. This disease has been associated with learning and memory deficits in children, whereas reduced attention levels, impaired work capacity, and cognitive deficits have been observed in adults. Strongly correlated with poverty and lack of basic sanitary conditions, this chronic endemic infection is common in Africa, South America, and parts of Asia and contributes to inhibition of social development and low quality of life in affected areas. Nonetheless, studies on the mechanisms involved in the neurological impairment caused by schistosomiasis are scarce. Here, we used a murine model of infection with Schistosoma mansoni in which parasites do not invade the central nervous system to evaluate the consequences of systemic infection on neurologic function. We observed that systemic infection with S. mansoni led to astrocyte and microglia activation, expression of oxidative stress-induced transcription factor Nrf2, oxidative damage, Tau phosphorylation, and amyloid-ß peptide accumulation in the prefrontal cortex of infected animals. We also found impairment in spatial learning and memory as evaluated by the Morris water maze task. Administration of anthelmintic (praziquantel) and antioxidant (N-acetylcysteine plus deferoxamine) treatments was effective in inhibiting most of these phenotypes, and the combination of both treatments had a synergistic effect to prevent such changes. These data demonstrate new perspectives toward the understanding of the pathology and possible therapeutic approaches to counteract long-term effects of systemic schistosomiasis on brain function.


Assuntos
Astrócitos/patologia , Microglia/patologia , Doenças Neurodegenerativas/patologia , Schistosoma mansoni/isolamento & purificação , Esquistossomose mansoni/complicações , Acetilcisteína/farmacologia , Animais , Anti-Helmínticos/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Desferroxamina/farmacologia , Modelos Animais de Doenças , Sequestradores de Radicais Livres/farmacologia , Masculino , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Teste do Labirinto Aquático de Morris/efeitos dos fármacos , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/etiologia , Praziquantel/farmacologia , Schistosoma mansoni/efeitos dos fármacos , Schistosoma mansoni/metabolismo , Esquistossomose mansoni/parasitologia , Esquistossomose mansoni/patologia , Sideróforos/farmacologia
10.
J Pharmacokinet Pharmacodyn ; 49(3): 293-310, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35024995

RESUMO

Racemic praziquantel (PZQ) is the standard treatment for schistosomiasis and liver fluke infections (opisthorchiasis and clonorchiasis). The development of an optimal pediatric formulation and dose selection would benefit from a population pharmacokinetic (popPK) model. A popPK model was developed for R-PZQ, the active enantiomer of PZQ, in 664 subjects, 493 African children (2-15 years) infected with Schistosoma mansoni and S. haematobium, and 171 Lao adults (15-78 years) infected with Opisthorchis viverrini. Racemate tablets were administered as single doses of 20, 40 and 60 mg/kg in children and 30, 40 and 50 mg/kg in 129 adults, and as 3 × 25 mg/kg apart in 42 adults. Samples collected by the dried-blood-spot technique were assayed by LC-MS/MS. A two-compartment disposition model, with allometric scaling and dual first-order and transit absorption, was developed using Phoenix™ software. Inversely parallel functions of age described the apparent oral bioavailability (BA) and clearance maturation in children and ageing in adults. BA decreased slightly in children with dose increase, and by 35% in adults with multiple dosing. Crushing tablets for preschool-aged children increased the first-order absorption rate by 64%. The mean transit absorption time was 70% higher in children. A popPK model for R-PZQ integrated African children over 2 years of age with schistosomiasis and Lao adults with opisthorchiasis, and should be useful to support dose optimization in children. In vitro hepatic and intestinal metabolism data would help refining and validating the model in younger children as well as in target ethnic pediatric and adult groups.


Assuntos
Anti-Helmínticos , Opistorquíase , Opisthorchis , Esquistossomose , Adulto , Animais , Anti-Helmínticos/farmacocinética , Anti-Helmínticos/uso terapêutico , Criança , Pré-Escolar , Cromatografia Líquida , Humanos , Laos , Opistorquíase/tratamento farmacológico , Opisthorchis/metabolismo , Praziquantel/farmacocinética , Praziquantel/uso terapêutico , Schistosoma mansoni/metabolismo , Esquistossomose/tratamento farmacológico , Espectrometria de Massas em Tandem
11.
Glycobiology ; 31(8): 1005-1017, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-33909073

RESUMO

Paucimannosidic glycans are restricted to the core structure [Man1-3GlcNAc2Fuc0-1] of N-glycans and are rarely found in mammalian tissues. Yet, especially [Man2-3GlcNAc2Fuc1] have been found significantly upregulated in tumors, including in colorectal and liver cancer. Mannitou IgM is a murine monoclonal antibody that was previously shown to recognize Man3GlcNAc2 with an almost exclusive selectivity. Here, we have sought the definition of the minimal glycan epitope of Mannitou IgM, initiated by screening on a newly designed paucimannosidic glycan microarray; among the best binders were Man3GlcNAc2 and its α1,6 core-fucosylated variant, Man3GlcNAc2Fuc1. Unexpectedly and in contrast to earlier findings, Man5GlcNAc2-type structures bind equally well and a large tolerance was observed for substitutions on the α1,6 arm. It was confirmed that any substitution on the single α1,3-linked mannose completely abolishes binding. Surface plasmon resonance for kinetic measurements of Mannitou IgM binding, either directly on the glycans or as presented on omega-1 and kappa-5 soluble egg antigens from the helminth parasite Schistosoma mansoni, showed submicromolar affinities. To characterize the epitope in greater and atomic detail, saturation transfer difference nuclear magnetic resonance spectroscopy was performed with the Mannitou antigen-binding fragment. The STD-NMR data demonstrated the strongest interactions with the aliphatic protons H1 and H2 of the α1-3-linked mannose and weaker imprints on its H3, H4 and H5 protons. In conclusion, Mannitou IgM binding requires a nonsubstituted α1,3-linked mannose branch of paucimannose also on proteins, making it a highly specific tool for the distinction of concurrent human tumor-associated carbohydrate antigens.


Assuntos
Glicoproteínas , Schistosoma mansoni , Animais , Proteínas de Ligação a DNA , Epitopos/química , Fucose/metabolismo , Glicoproteínas/metabolismo , Humanos , Imunoglobulina M , Mamíferos/metabolismo , Proteínas de Membrana , Camundongos , Polissacarídeos/química , Schistosoma mansoni/química , Schistosoma mansoni/metabolismo
12.
PLoS Pathog ; 15(9): e1007924, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31487324

RESUMO

Parasitic helminths evade, skew and dampen human immune responses through numerous mechanisms. Such effects will likely have consequences for HIV-1 transmission and disease progression. Here we analyzed the effects that soluble egg antigen (SEA) from Schistosoma mansoni had on modulating HIV-1 infection and cytokine/chemokine production in vitro. We determined that SEA, specifically through kappa-5, can potently bind to DC-SIGN and thereby blocks DC-SIGN mediated HIV-1 trans-infection (p<0.05) whilst not interfering with cis-infection. DCs exposed to SEA whilst maturing under Th2 promoting conditions, will upon co-culture with naïve T-cells induce a T-cell population that was less susceptible to HIV-1 R5 infection (p<0.05) compared to DCs unexposed to SEA, whereas HIV-1 X4 virus infection was unaffected. This was not observed for DCs exposed to SEA while maturing under Th1 or Th1/Th2 (Tmix) promoting conditions. All T-cell populations induced by SEA exposed DCs demonstrate a reduced capacity to produce IFN-γ and MIP-1ß. The infection profile of T-cells infected with HIV-1 R5 was not associated with down-modulation of CCR5 cell surface expression. We further show that DCs maturing under Tmix conditions exposed to plant recombinant omega-1 protein (rω-1), which demonstrates similar functions to natural ω-1, induced T-cell populations that were less sensitive for HIV-1 R5 infection (p<0.05), but not for X4 virus infection. This inhibition associated again with a reduction in IFN-γ and MIP-1ß expression, but additionally correlated with reduced CCR5 expression. We have shown that SEA parasite antigens and more specifically rω-1 can modulate HIV-1 infectivity with the potential to influence disease course in co-infected individuals.


Assuntos
Anticorpos Anti-Helmínticos/imunologia , Antígenos de Helmintos/imunologia , Proteínas do Ovo/imunologia , Infecções por HIV/metabolismo , Animais , Anticorpos Anti-Helmínticos/metabolismo , Antígenos de Helmintos/metabolismo , Linfócitos T CD4-Positivos/imunologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Proteínas do Ovo/metabolismo , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , HIV-1/imunologia , Humanos , Ativação Linfocitária , Receptores CCR5/metabolismo , Schistosoma mansoni/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Replicação Viral/imunologia
13.
Cytokine ; 138: 155390, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33341001

RESUMO

Morbidity during chronic schistosomiasis has been associated with the induction and modulation of type-2 granulomatous inflammatory response induced by antigens secreted by the eggs, which become trapped in capillary venules of the host tissues, especially in the liver and intestines. IL-33, an alarmin released after cell damage, binds to its ST2 (suppressor of tumorigenicity 2) receptor, expressed in an variety of immune cells, including ILC2 and macrophages, and stimulates the early production of IL-5 and IL-13, which leads to eosinophil infiltration and activation of a Th2 response. However, the role of IL-33/ST2 activation on Schistosoma-induced granuloma formation and modulation is mostly unknown. In the current work, we comparatively evaluated the immune response and granuloma formation in wild-type BALB/c (WT) and BALB/c mice genetically deficient in the IL-33 receptor (ST2-/-) experimentally infected with Schistosoma mansoni. Mice were infected with 25 or 50 S. mansoni cercariae and followed for up to 14 weeks to assess mortality. Mice from each experimental group were comparatively evaluated for parasite burden, liver immune response, and granuloma appearance during acute and chronic schistosomiasis. Our data showed that the number of circulating worms and eggs retained in the liver and eliminated in the feces was similar in WT and ST2-/- infected mice, but infected ST2-/- mice presented an enhanced rate of mortality. Interestingly, the production of type-2 cytokines by soluble egg antigens (SEA)-stimulated spleen cells, the serum concentrations of IL-5 and Immunoglobulin (Ig)-E, and the level of parasite-reactive IgG1 were similar in infected mice of both experimental groups. The concentrations of IL-4, IL-5, IL-13, and IFN-γ in liver homogenate of infected mice also did not differ between the strains at acute schistosomiasis, but there was a significant increase in IL-17 levels in ST2-/- infected mice at this phase. On the other hand, IL-4, IL-13, IL-10, IL-17, and IFN-γ concentrations were reduced and the ratios of IL-4/IFN-γ and IL-17/IFN-γ were higher in liver homogenate of chronically infected ST2-/- mice, suggesting unbalanced Th2 and Th17 responses. Moreover, liver granulomas of ST2-/- mice were larger and disorganized, showing an intense cellular infiltrate, rich in eosinophils and neutrophils. Our results suggest that the absence of the IL-33/ST2 pathway is not essential for the Schistosoma-induced Th2 response, but is necessary to prevent host mortality by modulating granuloma-mediated pathology.


Assuntos
Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Schistosoma mansoni/metabolismo , Células Th2/citologia , Animais , Antígenos/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Eritrócitos/metabolismo , Feminino , Granuloma/metabolismo , Sistema Imunitário , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/metabolismo , Esquistossomose mansoni , Baço/metabolismo , Células Th17/metabolismo
14.
Anal Bioanal Chem ; 413(10): 2755-2766, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33723627

RESUMO

Schistosoma mansoni is a parasitic flatworm causing schistosomiasis, an infectious disease affecting several hundred million people worldwide. Schistosomes live dioeciously, and upon pairing with the male, the female starts massive egg production, which causes pathology. Praziquantel (PZQ) is the only drug used, but it has an inherent risk of resistance development. Therefore, alternatives are needed. In the context of drug repurposing, the cancer drug imatinib was tested, showing high efficacy against S. mansoni in vitro. Besides the gonads, imatinib mainly affected the integrity of the intestine in males and females. In this study, we investigated the potential uptake and distribution of imatinib in adult schistosomes including its distribution kinetics. To this end, we applied for the first time atmospheric-pressure scanning microprobe matrix-assisted laser desorption/ionization mass spectrometry imaging (AP-SMALDI MSI) for drug imaging in paired S. mansoni. Our results indicate that imatinib was present in the esophagus and intestine of the male as early as 20 min after in vitro exposure, suggesting an oral uptake route. After one hour, the drug was also found inside the paired female. The detection of the main metabolite, N-desmethyl imatinib, indicated metabolization of the drug. Additionally, a marker signal for the female ovary was successfully applied to facilitate further conclusions regarding organ tropism of imatinib. Our results demonstrate that AP-SMALDI MSI is a useful method to study the uptake, tissue distribution, and metabolization of imatinib in S. mansoni. The results suggest using AP-SMALDI MSI also for investigating other antiparasitic compounds and their metabolites in schistosomes and other parasites.


Assuntos
Antineoplásicos/análise , Antiparasitários/análise , Mesilato de Imatinib/análise , Schistosoma mansoni/efeitos dos fármacos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Animais , Antineoplásicos/farmacocinética , Antiparasitários/farmacocinética , Reposicionamento de Medicamentos , Feminino , Masculino , Mesocricetus , Schistosoma mansoni/citologia , Schistosoma mansoni/metabolismo , Esquistossomose mansoni/tratamento farmacológico , Esquistossomose mansoni/parasitologia
15.
Nucleic Acids Res ; 47(11): 5973-5987, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31066441

RESUMO

Association of the initiation factor eIF4E with the mRNA cap structure is a key step for translation. Trypanosomatids present six eIF4E homologues, showing a low conservation and also differing significantly from the IF4Es of multicellular eukaryotes. On the mRNA side, while in most eukaryotes the mRNA contains cap-0 (7-methyl-GTP), the trypanosomatid mRNA features a cap-4, which is formed by a cap-0, followed by the AACU sequence containing 2'-O-ribose methylations and base methylations on nucleotides 1 and 4. The studies on eIF4E-cap-4 interaction have been hindered by the difficulty to synthesize this rather elaborated cap-4 sequence. To overcome this problem, we applied a liquid-phase oligonucleotide synthesis strategy and describe for the first time the crystal structure of a trypanosomatid eIF4E (T. cruzi EIF4E5) in complex with cap-4. The TcEIF4E5-cap-4 structure allowed a detailed description of the binding mechanism, revealing the interaction mode for the AACU sequence, with the bases packed in a parallel stacking conformation and involved, together with the methyl groups, in hydrophobic contacts with the protein. This binding mechanism evidences a distinct cap interaction mode in comparison with previously described eIF4E structures and may account for the difference of TcEIF4E5-cap-4 dissociation constant in comparison with other eIF4E homologues.


Assuntos
Fator de Iniciação 4E em Eucariotos/metabolismo , Capuzes de RNA/química , Trypanosoma cruzi/química , Animais , Proteínas de Transporte/metabolismo , Cristalografia por Raios X , Metilação de DNA , Humanos , Ligantes , Modelos Moleculares , Nucleotídeos/química , Oligonucleotídeos , Ligação Proteica , Análogos de Capuz de RNA/metabolismo , RNA Mensageiro/metabolismo , Schistosoma mansoni/metabolismo , Temperatura , Trypanosoma/metabolismo
16.
J Proteome Res ; 19(1): 314-326, 2020 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-31729880

RESUMO

Schistosomes are blood-dwelling helminth parasites that cause schistosomiasis, a debilitating disease resulting in inflammation and, in extreme cases, multiple organ damage. Major challenges to control the transmission persist, and the discovery of protective antigens remains of critical importance for vaccine development. Rhesus macaques can self-cure following schistosome infection, generating antibodies that target proteins from the tegument, gut, and esophagus, the last of which is the least investigated. We developed a dissection technique that permitted increased sensitivity in a comparative proteomics profiling of schistosome esophagus and gut. Proteome analysis of the male schistosome esophagus identified 13 proteins encoded by microexon genes (MEGs), 11 of which were uniquely located in the esophageal glands. Based on this and transcriptome information, a QconCAT was designed for the absolute quantification of selected targets. MEGs 12, 4.2, and 4.1 and venom allergen-like protein 7 were the most abundant, spanning over 245 million to 6 million copies per cell, while aspartyl protease, palmitoyl thioesterase, and galactosyl transferase were present at <1 million copies. Antigenic variation by alternative splicing of MEG proteins was confirmed together with a specialized machinery for protein glycosylation/secretion in the esophagus. Moreover, some gastrodermal secretions were highly enriched in the gut, while others were more uniformly distributed throughout the parasite, potentially indicating lysosomal activity. Collectively, our findings provide a more rational, better-oriented selection of schistosome vaccine candidates in the context of a proven model of protective immunity.


Assuntos
Trato Gastrointestinal/metabolismo , Proteínas de Helminto/metabolismo , Proteômica/métodos , Schistosoma mansoni/metabolismo , Animais , Esôfago/metabolismo , Ontologia Genética , Proteínas de Helminto/análise , Proteínas de Helminto/genética , Masculino , Camundongos , Schistosoma mansoni/patogenicidade , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
17.
PLoS Pathog ; 14(2): e1006870, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29425229

RESUMO

Protease inhibitors have important function during homeostasis, inflammation and tissue injury. In this study, we described the role of Schistosoma mansoni SmKI-1 serine protease inhibitor in parasite development and as a molecule capable of regulating different models of inflammatory diseases. First, we determine that recombinant (r) SmKI-1 and its Kunitz domain but not the C-terminal region possess inhibitory activity against trypsin and neutrophil elastase (NE). To better understand the molecular basis of NE inhibition by SmKI-1, molecular docking studies were also conducted. Docking results suggest a complete blockage of NE active site by SmKI-1 Kunitz domain. Additionally, rSmKI-1 markedly inhibited the capacity of NE to kill schistosomes. In order to further investigate the role of SmKI-1 in the parasite, we designed specific siRNA to knockdown SmKI-1 in S. mansoni. SmKI-1 gene suppression in larval stage of S. mansoni robustly impact in parasite development in vitro and in vivo. To determine the ability of SmKI-1 to interfere with neutrophil migration and function, we tested SmKI-1 anti-inflammatory potential in different murine models of inflammatory diseases. Treatment with SmKI-1 rescued acetaminophen (APAP)-mediated liver damage, with a significant reduction in both neutrophil recruitment and elastase activity. In the model of gout arthritis, this protein reduced neutrophil accumulation, IL-1ß secretion, hypernociception, and overall pathological score. Finally, we demonstrated the ability of SmKI-1 to inhibit early events that trigger neutrophil recruitment in pleural cavities of mice in response to carrageenan. In conclusion, SmKI-1 is a key protein in S. mansoni survival and it has the ability to inhibit neutrophil function as a promising therapeutic molecule against inflammatory diseases.


Assuntos
Inflamação/metabolismo , Elastase de Leucócito/metabolismo , Neutrófilos/efeitos dos fármacos , Schistosoma mansoni , Inibidores de Serina Proteinase/metabolismo , Inibidores de Serina Proteinase/farmacologia , Animais , Células Cultivadas , Feminino , Inflamação/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Simulação de Acoplamento Molecular , Neutrófilos/fisiologia , Ligação Proteica , Schistosoma mansoni/imunologia , Schistosoma mansoni/metabolismo , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/metabolismo
18.
PLoS Pathog ; 14(5): e1007066, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29782530

RESUMO

Epigenetic mechanisms and chromatin structure play an important role in development. Their impact is therefore expected to be strong in parasites with complex life cycles and multiple, strikingly different, developmental stages, i.e. developmental plasticity. Some studies have already described how the chromatin structure, through histone modifications, varies from a developmental stage to another in a few unicellular parasites. While H3K4me3 profiles remain relatively constant, H3K27 trimethylation and bivalent methylation show strong variation. Inhibitors (A366 and GSK343) of H3K27 histone methyltransferase activity in S. mansoni efficiently blocked miracidium to sporocyst transition indicating that H3K27 trimethylation is required for life cycle progression. As S. mansoni is a multicellular parasite that significantly affects both the health and economy of endemic areas, a better understanding of fluke developmental processes within the definitive host will likely highlight novel disease control strategies. Towards this goal, we also studied H4K20me1 in female cercariae and adults. In particular, we found that bivalent trimethylation of H3K4 and H3K27 at the transcription start site of genes is a landmark of the cercarial stage. In cercariae, H3K27me3 presence and strong enrichment in H4K20me1 over long regions (10-100 kb) is associated with development related genes. Here, we provide a broad overview of the chromatin structure of a metazoan parasite throughout its most important lifecycle stages. The five developmental stages studied here present distinct chromatin structures, indicating that histone methylation plays an important role during development. Hence, components of the histone methylation (and demethylation) machinery may provide suitable Schistosomiasis control targets.


Assuntos
Biomphalaria/parasitologia , Histonas/metabolismo , Estágios do Ciclo de Vida/fisiologia , Schistosoma mansoni/metabolismo , Esquistossomose mansoni/parasitologia , Animais , Cromatina/química , Imunoprecipitação da Cromatina , Cricetinae , Feminino , Água Doce , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histonas/química , Histonas/genética , Humanos , Fígado/parasitologia , Masculino , Metilação , Camundongos , Sequências Repetitivas de Ácido Nucleico/genética , Schistosoma mansoni/genética , Schistosoma mansoni/crescimento & desenvolvimento , Alinhamento de Sequência
19.
PLoS Pathog ; 14(1): e1006718, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29346437

RESUMO

Schistosomes are blood-dwelling trematodes with global impact on human and animal health. Because medical treatment is currently based on a single drug, praziquantel, there is urgent need for the development of alternative control strategies. The Schistosoma mansoni genome project provides a platform to study and connect the genetic repertoire of schistosomes to specific biological functions essential for successful parasitism. G protein-coupled receptors (GPCRs) form the largest superfamily of transmembrane receptors throughout the Eumetazoan phyla, including platyhelminths. Due to their involvement in diverse biological processes, their pharmacological importance, and proven druggability, GPCRs are promising targets for new anthelmintics. However, to identify candidate receptors, a more detailed understanding of the roles of GPCR signalling in schistosome biology is essential. An updated phylogenetic analysis of the S. mansoni GPCR genome (GPCRome) is presented, facilitated by updated genome data that allowed a more precise annotation of GPCRs. Additionally, we review the current knowledge on GPCR signalling in this parasite and provide new insights into the potential roles of GPCRs in schistosome reproduction based on the findings of a recent tissue-specific transcriptomic study in paired and unpaired S. mansoni. According to the current analysis, GPCRs contribute to gonad-specific functions but also to nongonad, pairing-dependent processes. The latter may regulate gonad-unrelated functions during the multifaceted male-female interaction. Finally, we compare the schistosome GPCRome to that of another parasitic trematode, Fasciola, and discuss the importance of GPCRs to basic and applied research. Phylogenetic analyses display GPCR diversity in free-living and parasitic platyhelminths and suggest diverse functions in schistosomes. Although their roles need to be substantiated by functional studies in the future, the data support the selection of GPCR candidates for basic and applied studies, invigorating the exploitation of this important receptor class for drug discovery against schistosomes but also other trematodes.


Assuntos
Quinases de Receptores Acoplados a Proteína G/metabolismo , Proteínas de Helminto/metabolismo , Modelos Biológicos , Schistosoma mansoni/metabolismo , Transdução de Sinais , Animais , Antiplatelmínticos/farmacologia , Fasciola/efeitos dos fármacos , Fasciola/genética , Fasciola/metabolismo , Fasciola/patogenicidade , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/química , Quinases de Receptores Acoplados a Proteína G/genética , Perfilação da Expressão Gênica , Genoma Helmíntico , Genômica/métodos , Proteínas de Helminto/antagonistas & inibidores , Proteínas de Helminto/química , Proteínas de Helminto/genética , Humanos , Especificidade de Órgãos , Filogenia , Inibidores de Proteínas Quinases/farmacologia , Schistosoma mansoni/efeitos dos fármacos , Schistosoma mansoni/genética , Schistosoma mansoni/patogenicidade , Transdução de Sinais/efeitos dos fármacos
20.
Parasitology ; 147(8): 865-872, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31840628

RESUMO

Schistosomiasis is an acute and chronic disease caused by parasitic worms of the genus Schistosoma. Treatment is solely dependent on praziquantel. In the face of the worldwide dimension, projects have been initiated to develop new chemotherapies. Due to their proven druggability, G protein-coupled receptors (GPCRs) are promising targets for anthelmintics. However, to identify candidate receptors, a deeper understanding of GPCR signalling in schistosome biology is essential. Comparative transcriptomics of paired and unpaired worms and their gonads revealed 59 differentially regulated GPCR-coding genes putatively involved in neuronal processes. In general, the diversity among GPCRs and their integral membrane topology make it difficult to characterize and deorphanize these receptors. To overcome existing limitations, we performed a pilot approach and utilized the innovative Membrane-Anchored Ligand And Receptor yeast two-hybrid system (MALAR-Y2H) to associate potential neuropeptide ligands with their cognate receptors. Here, we demonstrated the ability to express full-length GPCRs of Schistosoma mansoni in a heterologous yeast-based system. Additionally, we localized GPCRs and chimeras of neuropeptides fused to the WBP1 transmembrane domain of yeast to the plasma membrane of yeast cells. Reporter gene assays indicated ligand-receptor binding, which allowed us to identify certain neuropeptides as potential ligands for two GPCRs, which had been found before to be differentially expressed in schistosomes in a pairing-dependent manner. Thus, the MALAR-Y2H system appears suitable to unravel schistosome GPCR-ligand interactions. Besides its relevance for understanding schistosome biology, identifying and characterizing GPCR-ligand interaction will also contribute to applied research aspects.


Assuntos
Neuropeptídeos/metabolismo , Receptores Acoplados a Proteínas G , Schistosoma mansoni , Técnicas do Sistema de Duplo-Híbrido , Animais , Proteínas de Ligação ao GTP/metabolismo , Humanos , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Schistosoma mansoni/genética , Schistosoma mansoni/metabolismo , Transformação Genética , Leveduras/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA