Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 326
Filtrar
1.
J Neuroimmune Pharmacol ; 19(1): 37, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052165

RESUMEN

Recombinant interleukin-22 (rIL-22) has been reported as a protective agent in murine models of diseases driven by epithelial injury. Parasites have a circadian rhythm and their sensitivity to a certain drug may vary during the day. Therefore, this work aimed to investigate the effect of rIL-22 administration at different times of the day on the inflammation, oxidative status, and neurotransmitter release in the gut-brain axis of the Schistosoma mansoni-infected mice. Sixty male BALB/c mice aged six weeks weighing 25-30 g were divided into a control group (injected intraperitoneally with PBS), mice infected with 80 ± 10 cercariae of S. mansoni (infected group) then injected intraperitoneally with PBS, and rIL-22 treated groups. rIL-22 was administrated intraperitoneally (400 ng/kg) either at the onset or offset of the light phase for 14 days. IL-22 administration reduced the levels of IL-1ß, tumor necrosis factor-alpha (TNF-α), nuclear factor kappa beta (NF-κß), and enhanced the production of IL-22 and IL-17. The treatment with IL-22 increased glutathione (GSH) and reduced malondialdehyde (MDA) and nitric oxide (NO) levels both in the ileum and brain. The B-cell lymphoma 2 (BCL2) protein level in the ileum was diminished after IL-22 administration. Brain-derived neurotrophic factor (BDNF) and neurotransmitter release (serotonin, 5HT, norepinephrine, NE, dopamine, DA, Glutamate, Glu, and -amino butyric acid, GABA) were improved by rIL-22. In conclusion, rIL-22 showed promising immunotherapy for inflammation, oxidative damage, and neuropathological signs associated with schistosomiasis. The efficacy of IL-22 increased significantly upon its administration at the time of light offset.


Asunto(s)
Eje Cerebro-Intestino , Interleucina-22 , Interleucinas , Ratones Endogámicos BALB C , Neurotransmisores , Proteínas Recombinantes , Esquistosomiasis mansoni , Animales , Ratones , Masculino , Neurotransmisores/metabolismo , Neurotransmisores/farmacología , Interleucinas/metabolismo , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/administración & dosificación , Eje Cerebro-Intestino/efectos de los fármacos , Eje Cerebro-Intestino/fisiología , Inmunoterapia/métodos , Monoaminas Biogénicas/metabolismo , Inflamación/metabolismo , Inflamación/tratamiento farmacológico
2.
BMC Infect Dis ; 24(1): 636, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38918706

RESUMEN

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.


Asunto(s)
Mononucleótido de Flavina , Flavina-Adenina Dinucleótido , Riboflavina , Schistosoma mansoni , Riboflavina/metabolismo , Mononucleótido de Flavina/metabolismo , Animales , Flavina-Adenina Dinucleótido/metabolismo , Schistosoma mansoni/metabolismo , Schistosoma mansoni/genética , Ratones , Humanos , Esquistosomiasis mansoni/parasitología , Esquistosomiasis mansoni/metabolismo
3.
Front Immunol ; 15: 1372927, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38742105

RESUMEN

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.


Asunto(s)
Dinoprostona , Lectinas Tipo C , Manosa , Polisacáridos , Schistosoma mansoni , Células Th2 , Animales , Ratones , Antígenos Helmínticos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Dinoprostona/metabolismo , Lectinas Tipo C/metabolismo , Lectinas Tipo C/inmunología , Manosa/metabolismo , Manosa/inmunología , Ratones Endogámicos C57BL , Óvulo/inmunología , Óvulo/metabolismo , Ligando OX40/metabolismo , Polisacáridos/inmunología , Polisacáridos/metabolismo , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/metabolismo , Esquistosomiasis mansoni/parasitología , Células Th2/inmunología , Células Th2/metabolismo
4.
Anal Chem ; 96(16): 6311-6320, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38594017

RESUMEN

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.


Asunto(s)
Glicoesfingolípidos , Hígado , Schistosoma mansoni , Esquistosomiasis mansoni , Animales , Glicoesfingolípidos/metabolismo , Glicoesfingolípidos/química , Hígado/metabolismo , Hígado/parasitología , Cricetinae , Esquistosomiasis mansoni/parasitología , Esquistosomiasis mansoni/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrometría de Masas en Tándem , Mesocricetus , Cromatografía Liquida , Masculino
5.
Cell Mol Gastroenterol Hepatol ; 17(1): 107-117, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37696392

RESUMEN

BACKGROUND & AIMS: Schistosomiasis is one of the most prominent parasite-induced infectious diseases, affecting more than 250 million people. Schistosoma mansoni causes metabolic exhaustion and a strong redox imbalance in the liver, causing parenchymal damage, and may predispose for cancer. We investigated whether oxidative stress provokes hepatocellular proliferation upon S. mansoni infection. METHODS: The cell cycle, replication stress response, and proliferation were analyzed on transcriptional and protein levels in the livers of S. mansoni-infected hamsters and by mechanistic gain- and loss-of-function experiments in human hepatoma cells. Major results were validated in human biopsy specimens of S. mansoni-infected patients. RESULTS: S. mansoni infection induced licensing factors of DNA replication and cell-cycle checkpoint cyclins in parallel with a DNA damage response in hamster hepatocytes. Moreover, even unisexual infection without egg effects, as a reflection of a chronic inflammatory process, resulted in a moderate activation of several cell-cycle markers. S. mansoni soluble egg antigens induced proliferation of human hepatoma cells that could be abolished by reduced glutathione. CONCLUSIONS: Our data suggest that hepatocellular proliferation is triggered by S. mansoni egg-induced oxidative stress.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Esquistosomiasis mansoni , Cricetinae , Animales , Humanos , Schistosoma mansoni/fisiología , Esquistosomiasis mansoni/metabolismo , Estrés Oxidativo , Proliferación Celular
6.
Sci Data ; 10(1): 775, 2023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37935722

RESUMEN

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.


Asunto(s)
Schistosoma mansoni , Transcriptoma , Animales , Femenino , Humanos , Masculino , Perfilación de la Expresión Génica , Schistosoma mansoni/genética , Esquistosomiasis mansoni/metabolismo , Esquistosomiasis mansoni/parasitología , Factores Sexuales
7.
Front Immunol ; 13: 956871, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36131936

RESUMEN

The freshwater snail Biomphalaria glabrata is an intermediate host of Schistosoma mansoni, the agent of human intestinal schistosomiasis. However, much is to be discovered about its innate immune system that appears as a complex black box, in which the immune cells (called hemocytes) play a major role in both cellular and humoral response towards pathogens. Until now, hemocyte classification has been based exclusively on cell morphology and ultrastructural description and depending on the authors considered from 2 to 5 hemocyte populations have been described. In this study, we proposed to evaluate the hemocyte heterogeneity at the transcriptomic level. To accomplish this objective, we used single cell RNA sequencing (scRNAseq) technology coupled to a droplet-based system to separate hemocytes and analyze their transcriptome at a unique cell level in naive Biomphalaria glabrata snails. We were able to demonstrate the presence of 7 hemocyte transcriptomic populations defined by the expression of specific marker genes. As a result, scRNAseq approach showed a high heterogeneity within hemocytes, but provides a detailed description of the different hemocyte transcriptomic populations in B. glabrata supported by distinct cellular functions and lineage trajectory. As a main result, scRNAseq revealed the 3 main population as a super-group of hemocyte diversity but, on the contrary, a great hemocytes plasticity with a probable capacity of hemocytes to engage to different activation pathways. This work opens a new field of research to understand the role of hemocytes particularly in response to pathogens, and towards S. mansoni parasites.


Asunto(s)
Biomphalaria , Esquistosomiasis mansoni , Animales , Biomphalaria/parasitología , Hemocitos , Humanos , Schistosoma mansoni , Esquistosomiasis mansoni/metabolismo , Análisis de Secuencia de ARN , Caracoles
8.
Med Microbiol Immunol ; 211(4): 211-218, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35819523

RESUMEN

Schistosoma mansoni infections, particularly egg antigens, induce Th2-dominant granulomatous responses accompanied by remarkable immunoregulatory mechanisms that avoid intense fibrosis. Interleukin (IL)-33 is a cytokine that stimulates the early activation of Th2 responses, and its soluble ST2 receptor (sST2) avoids granulomatous response, as well as CXCL9 and CXCL10 chemokines that have antifibrotic activity. However, in schistosomiasis, these molecules have not been suitably studied. Therefore, this study aimed to measure IL-33 and sST2 RNA, cytokines, and chemokines in peripheral blood cultures from individuals living in schistosomiasis-endemic areas. Peripheral blood cells from individuals with S. mansoni (n = 34) and non-infected individuals (n = 31) were cultured under mitogen stimulation. Supernatant chemokines and cytokines were evaluated using a cytometric bead array, and IL-33 and sST2 mRNA expression was measured using qPCR. Infected individuals showed higher levels of CXCL8, CXCL9, CXCL10, IFN-γ, TNF-α, IL-6, IL-2, IL-4, and IL-10; there was a lower expression of IL-33 mRNA and similar expression of sST2mRNA in infected than non-infected individuals. In conclusion, for the first time, we demonstrated lower IL-33mRNA expression and high levels of the antifibrotic chemokines CXCL9 and CXCL10 in schistosomiasis mansoni, which could control exacerbations of the disease in individuals from endemic areas.


Asunto(s)
Esquistosomiasis mansoni , Esquistosomiasis , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Humanos , Interleucina-33/metabolismo , Leucocitos Mononucleares , ARN Mensajero , Esquistosomiasis/metabolismo , Esquistosomiasis mansoni/epidemiología , Esquistosomiasis mansoni/metabolismo
9.
Biochimie ; 194: 108-117, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34990770

RESUMEN

Schistosomiasis is a neglected tropical disease caused by parasitic flatworms of the genus Schistosoma. Mono-therapeutic treatment of this disease with the drug praziquantel, presents challenges such as inactivity against immature worms and inability to prevent reinfection. Importantly, ion channels are important targets for many current anthelmintics. Transient receptor potential (TRP) channels are important mediators of sensory signals with marked effects on cellular functions and signaling pathways. TRPML channels are a class of Ca2+-permeable TRP channels expressed on endolysosomal membranes. They regulate lysosomal function and trafficking, among other functions. Schistosoma mansoni is predicted to have a single TRPML gene (SmTRPML) with two splice variants differing by 12 amino acids. This study focuses on exploring the physiological properties of SmTRPML channels to better understand their role in schistosomes. In mammalian cells expressing SmTRPML, TRPML activators elicit a rise in intracellular Ca2+. In these cells, SmTRPML localizes both to lysosomes and the plasma membrane. These same TRPML activators elicit an increase in adult worm motility that is dependent on SmTRPML expression, indicating a role for these channels in parasite neuromuscular activity. Suppression of SmTRPML in adult worms, or exposure of adult worms to TRPML inhibitors, results in tegumental vacuolations, balloon-like surface exudates, and membrane blebbing, similar to that found following TRPML loss in other organisms. Together, these findings indicate that SmTRPML may regulate the function of the schistosome endolysosomal system. Further, the role of SmTRPML in neuromuscular activity and in parasite tegumental integrity establishes this channel as a candidate anti-schistosome drug target.


Asunto(s)
Antihelmínticos , Esquistosomiasis mansoni , Canales de Potencial de Receptor Transitorio , Animales , Antihelmínticos/metabolismo , Antihelmínticos/farmacología , Antihelmínticos/uso terapéutico , Endosomas/metabolismo , Praziquantel/metabolismo , Praziquantel/farmacología , Praziquantel/uso terapéutico , Schistosoma mansoni/metabolismo , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/metabolismo , Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo
10.
PLoS Negl Trop Dis ; 15(11): e0009898, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34731172

RESUMEN

Given the worldwide burden of neglected tropical diseases, there is ongoing need to develop novel anthelmintic agents to strengthen the pipeline of drugs to combat these burdensome infections. Many diseases caused by parasitic flatworms are treated using the anthelmintic drug praziquantel (PZQ), employed for decades as the key clinical agent to treat schistosomiasis. PZQ activates a flatworm transient receptor potential (TRP) channel within the melastatin family (TRPMPZQ) to mediate sustained Ca2+ influx and worm paralysis. As a druggable target present in many parasitic flatworms, TRPMPZQ is a promising target for a target-based screening campaign with the goal of discovering novel regulators of this channel complex. Here, we have optimized methods to miniaturize a Ca2+-based reporter assay for Schistosoma mansoni TRPMPZQ (Sm.TRPMPZQ) activity enabling a high throughput screening (HTS) approach. This methodology will enable further HTS efforts against Sm.TRPMPZQ as well as other flatworm ion channels. A pilot screen of ~16,000 compounds yielded a novel activator of Sm.TRPMPZQ, and numerous potential blockers. The new activator of Sm.TRPMPZQ represented a distinct chemotype to PZQ, but is a known chemical entity previously identified by phenotypic screening. The fact that a compound prioritized from a phenotypic screening campaign is revealed to act, like PZQ, as an Sm.TRPMPZQ agonist underscores the validity of TRPMPZQ as a druggable target for antischistosomal ligands.


Asunto(s)
Antihelmínticos/farmacología , Proteínas del Helminto/antagonistas & inhibidores , Praziquantel/farmacología , Schistosoma mansoni/metabolismo , Esquistosomiasis mansoni/parasitología , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Animales , Antihelmínticos/química , Calcio/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Humanos , Masculino , Ratones , Praziquantel/química , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/genética , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/metabolismo , Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo
11.
Immunohorizons ; 5(8): 721-732, 2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34462311

RESUMEN

Plasmacytoid dendritic cells (pDCs) are potent producers of type I IFN (IFN-I) during viral infection and respond to IFN-I in a positive feedback loop that promotes their function. IFN-I shapes dendritic cell responses during helminth infection, impacting their ability to support Th2 responses. However, the role of pDCs in type 2 inflammation is unclear. Previous studies have shown that pDCs are dispensable for hepatic or splenic Th2 responses during the early stages of murine infection with the trematode Schistosoma mansoni at the onset of parasite egg laying. However, during S. mansoni infection, an ongoing Th2 response against mature parasite eggs is required to protect the liver and intestine from acute damage and how pDCs participate in immune responses to eggs and adult worms in various tissues beyond acute infection remains unclear. We now show that pDCs are required for optimal Th2 cytokine production in response to S. mansoni eggs in the intestinal-draining mesenteric lymph nodes throughout infection and for egg-specific IFN-γ at later time points of infection. Further, pDC depletion at chronic stages of infection led to increased hepatic and splenic pathology as well as abrogated Th2 cell cytokine production and activation in the liver. In vitro, mesenteric lymph node pDCs supported Th2 cell responses from infection-experienced CD4+ T cells, a process dependent on pDC IFN-I responsiveness, yet independent of Ag. Together, these data highlight a previously unappreciated role for pDCs and IFN-I in maintaining and reinforcing type 2 immunity in the lymph nodes and inflamed tissue during helminth infection.


Asunto(s)
Citocinas/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos/inmunología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/parasitología , Citocinas/metabolismo , Células Dendríticas/parasitología , Femenino , Citometría de Flujo/métodos , Interacciones Huésped-Parásitos/inmunología , Recuento de Linfocitos , Ratones Endogámicos C57BL , Ratones Noqueados , Schistosoma mansoni/fisiología , Esquistosomiasis mansoni/metabolismo , Esquistosomiasis mansoni/parasitología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Colaboradores-Inductores/parasitología , Células Th2/inmunología , Células Th2/metabolismo , Células Th2/parasitología
12.
Mol Biotechnol ; 63(11): 983-991, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34165770

RESUMEN

Schistosomiasis causes significant morbidity and mortality. Vaccine efforts to date indicate the need to increase the immunogenicity of Schistosoma antigens. The multiple antigen-presenting system, whereby proteins are genetically fused to rhizavidin and affinity linked to biotinylated templates, enables the generation of robust immune responses. The objective of this work was to express and purify the S. mansoni antigens, SmTSP-2 and SmCD59.2, in fusion with rhizavidin. The fusion with rhizavidin greatly decreased the expression level of rSmTSP-2, but not rSmCD59.2, and both were expressed in the insoluble fraction, requiring optimization of culture conditions. Evaluation of different E. coli strains and media showed that BL21-DE3 cultured in Terrific Broth provided the highest expression levels of both proteins. Investigation of a range of time and temperature of induction showed that E. coli strains expressing rRzv:SmTSP-2 and rRzv:SmCD59.2 showed the highest protein production at 23 °C for 15 h. Recombinant proteins were purified by a single step of affinity chromatography allowing isolation of these proteins in high concentration and purity. The optimization process increased final soluble protein yield of rRzv:SmTSP-2 by fourfold and rRzv:SmCD59.2 by tenfold, providing ~ 20 mg/L of each protein. Optimized fusion protein production will allow antigen use in biotin-rhizavidin affinity platforms.


Asunto(s)
Antígenos Helmínticos/biosíntesis , Proteínas Bacterianas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Schistosoma mansoni/metabolismo , Esquistosomiasis mansoni/inmunología , Animales , Antígenos Helmínticos/genética , Antígenos Helmínticos/aislamiento & purificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Cromatografía de Afinidad/métodos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Schistosoma mansoni/química , Schistosoma mansoni/inmunología , Schistosoma mansoni/aislamiento & purificación , Esquistosomiasis mansoni/metabolismo , Esquistosomiasis mansoni/parasitología
13.
Artículo en Inglés | MEDLINE | ID: mdl-34119651

RESUMEN

Schistosoma mansoni worms are under a milieu of external and internal signaling pathways. The life-cycle stages are exposed to enormous stimuli within the mammalian and the snail hosts and as free-living stages in the fresh water. Furthermore, there is a unique interplay between the male and the female worms involving many stimuli from the male essential for full development of the female. PI3K/Akt/mTOR is an evolutionarily divergent signal transduction pathway universal to nearly every multicellular organism. This work reviews the Schistosoma mansoni PI3K/Akt/mTOR signal pathways and the involvement of the signal in the worms' physiology concerning the uptake of glucose, reproduction and survival. The inhibitors of the signal pathway used against Schistosoma mansoni were summarized. Given the importance of the PI3K/Akt/mTOR signal pathway, its inhibition could be a promising control strategy against schistosomiasis.


Asunto(s)
Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Schistosoma mansoni/fisiología , Esquistosomiasis mansoni/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Humanos , Esquistosomiasis mansoni/parasitología , Transducción de Señal
14.
J Comp Neurol ; 529(13): 3336-3358, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34041754

RESUMEN

Freshwater snails of the genus Biomphalaria serve as intermediate hosts for the digenetic trematode Schistosoma mansoni, the etiological agent for the most widespread form of intestinal schistosomiasis. As neuropeptide signaling in host snails can be altered by trematode infection, a neural transcriptomics approach was undertaken to identify peptide precursors in Biomphalaria glabrata, the major intermediate host for S. mansoni in the Western Hemisphere. Three transcripts that encode peptides belonging to the FMRF-NH2 -related peptide (FaRP) family were identified in B. glabrata. One transcript encoded a precursor polypeptide (Bgl-FaRP1; 292 amino acids) that included eight copies of the tetrapeptide FMRF-NH2 and single copies of FIRF-NH2 , FLRF-NH2 , and pQFYRI-NH2 . The second transcript encoded a precursor (Bgl-FaRP2; 347 amino acids) that comprised 14 copies of the heptapeptide GDPFLRF-NH2 and 1 copy of SKPYMRF-NH2 . The precursor encoded by the third transcript (Bgl-FaRP3; 287 amino acids) recapitulated Bgl-FaRP2 but lacked the full SKPYMRF-NH2 peptide. The three precursors shared a common signal peptide, suggesting a genomic organization described previously in gastropods. Immunohistochemical studies were performed on the nervous systems of B. glabrata and B. alexandrina, a major intermediate host for S. mansoni in Egypt. FMRF-NH2 -like immunoreactive (FMRF-NH2 -li) neurons were located in regions of the central nervous system associated with reproduction, feeding, and cardiorespiration. Antisera raised against non-FMRF-NH2 peptides present in the tetrapeptide and heptapeptide precursors labeled independent subsets of the FMRF-NH2 -li neurons. This study supports the participation of FMRF-NH2 -related neuropeptides in the regulation of vital physiological and behavioral systems that are altered by parasitism in Biomphalaria.


Asunto(s)
FMRFamida/genética , Neuropéptidos/genética , Esquistosomiasis mansoni/genética , Transcriptoma/genética , Secuencia de Aminoácidos , Animales , Biomphalaria , FMRFamida/análisis , FMRFamida/metabolismo , Neuropéptidos/análisis , Neuropéptidos/metabolismo , Imagen Óptica/métodos , Schistosoma mansoni/genética , Schistosoma mansoni/aislamiento & purificación , Esquistosomiasis mansoni/metabolismo
15.
Front Immunol ; 12: 624191, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33777004

RESUMEN

In spite of several decades of research, an effective vaccine against schistosomiasis remains elusive. The radiation-attenuated (RA) cercarial vaccine is still the best model eliciting high protection levels, although the immune mechanisms have not yet been fully characterized. In order to identify genes and pathways underlying protection we investigated patterns of gene expression in PBMC and skin draining Lymph Nodes (LN) from mice using two exposure comparisons: vaccination with 500 attenuated cercariae versus infection with 500 normal cercariae; one versus three doses. Vaccinated mice were challenged with 120 normal parasites. Integration of PBMC and LN data from the infected group revealed early up-regulation of pathways associated with Th2 skewing and polarization of IgG antibody profiles. Additionally, hemostasis pathways were downregulated in infected mice, correlating with platelet reduction, potentially a mechanism to assist parasite migration through capillary beds. Conversely, up regulation of such mechanisms after vaccination may explain parasite blockade in the lungs. In contrast, a single exposure to attenuated parasites revealed early establishment of a Th1 bias (signaling of IL-1, IFN-γ; and Leishmania infection). Genes encoding chemokines and their receptors were more prominent in vaccinated mice, indicating an enhanced capacity for inflammation, potentially augmenting the inhibition of intravascular migration. Increasing the vaccinations from one to three did not dramatically elevate protection, but there was a clear shift towards antibody-mediated effectors. However, elements of the Th1 bias were still evident. Notable features after three vaccinations were markers of cytotoxicity (including IL-6 and NK cells) together with growth factors and their receptors (FGFR/VEGF/EGF) and the apoptosis pathway. Indeed, there is evidence for the development of anergy after three vaccinations, borne out by the limited responses detected in samples after challenge. We infer that persistence of a Th1 response puts a limit on expression of antibody-mediated mechanisms. This feature may explain the failure of multiple doses to drive protection towards sterile immunity. We suggest that the secretions of lung stage parasites would make a novel cohort of antigens for testing in protection experiments.


Asunto(s)
Hemostasis , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Vacunas Antiprotozoos/administración & dosificación , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/prevención & control , Biología de Sistemas , Animales , Cercarias/inmunología , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Hemostasis/genética , Interacciones Huésped-Parásitos , Péptidos y Proteínas de Señalización Intercelular/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/parasitología , Ratones Endogámicos C57BL , Análisis por Micromatrices , Vacunas Antiprotozoos/inmunología , Schistosoma mansoni/patogenicidad , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/metabolismo , Esquistosomiasis mansoni/parasitología , Células TH1/inmunología , Células TH1/metabolismo , Células TH1/parasitología , Balance Th1 - Th2 , Células Th2/inmunología , Células Th2/metabolismo , Células Th2/parasitología , Factores de Tiempo , Transcriptoma , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología
16.
Sci Rep ; 11(1): 4476, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33627744

RESUMEN

Schistosoma mansoni is less susceptible to the antiparasitic drug ivermectin than other helminths. By inhibiting the P-glycoprotein or cytochrome P450 3A in mice host or parasites in a murine model, we aimed at increasing the sensitivity of S. mansoni to the drug and thus preventing infection. We assigned 124 BALB/c mice to no treatment, treatment with ivermectin only or a combination of ivermectin with either cobicistat or elacridar once daily for three days before infecting them with 150 S. mansoni cercariae each. The assignment was done by batches without an explicit randomization code. Toxicity was monitored. At eight weeks post-infection, mice were euthanized. We determined number of eggs in intestine and liver, adult worms in portal and mesenteric veins. Disease was assessed by counting granulomas/cm2 of liver and studying organ weight indices and total weight. IgG levels in serum were also considered. No difference between groups treated with ivermectin only or in combination with cobicistat or elacridar compared with untreated, infected controls. Most mice treated with ivermectin and elacridar suffered severe neurological toxicity. In conclusion, systemic treatment with ivermectin, even in the presence of pharmacological inhibition of P-glycoprotein or cytochrome P450 3A, did not result in effective prophylaxis for S. mansoni infection in an experimental murine model.


Asunto(s)
Acridinas/farmacología , Cobicistat/farmacología , Ivermectina/farmacología , Schistosoma mansoni/efectos de los fármacos , Esquistosomiasis mansoni/tratamiento farmacológico , Tetrahidroisoquinolinas/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Antiparasitarios/farmacología , Citocromo P-450 CYP3A/metabolismo , Femenino , Granuloma/tratamiento farmacológico , Granuloma/parasitología , Inmunoglobulina G/metabolismo , Intestinos/parasitología , Hígado/parasitología , Masculino , Venas Mesentéricas/metabolismo , Venas Mesentéricas/parasitología , Ratones , Ratones Endogámicos BALB C , Recuento de Huevos de Parásitos/métodos , Esquistosomiasis mansoni/metabolismo
17.
Int J Biol Macromol ; 175: 406-421, 2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33549669

RESUMEN

Schistosomiasis is a neglected tropical disease that affects more than 250 million people worldwide. The only drug available for its treatment undergoes first-pass hepatic metabolism and is not capable of preventing reinfection, which makes the search of new therapies urgently needed. Due to the essential role of fumarases in metabolism, these enzymes represent potential targets for developing novel schistosomiasis treatments. Here, we evaluate the expression profiles for class I and class II fumarases from Schistosoma mansoni (SmFHI and SmFHII, respectively), and report the complete characterization of SmFHII. The first SmFHII structure in complex with L-malate was determined at 1.85 Å resolution. The significant thermoshift observed for SmFHII in the presence of identified ligands makes the differential scanning fluorimetry an adequate technique for ligand screening. A complete kinetic characterization of SmFHII was performed, and comparison with the human fumarase (HsFH) revealed differences regarding the turnover number (kcat). Structural characterization allowed us to identify differences between SmFHII and HsFH that could be explored to design new selective inhibitors. This work represents the very first step towards validate the fumarases as drug targets to treat schistosomiasis. Our results provide the structural basis to rational search for selective ligands.


Asunto(s)
Fumarato Hidratasa/farmacología , Schistosoma mansoni/enzimología , Esquistosomiasis mansoni/tratamiento farmacológico , Animales , Femenino , Fumarato Hidratasa/metabolismo , Cinética , Ligandos , Masculino , Ratones , Schistosoma mansoni/metabolismo , Esquistosomiasis/tratamiento farmacológico , Esquistosomiasis/metabolismo , Esquistosomiasis mansoni/metabolismo
18.
PLoS Pathog ; 17(1): e1009198, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33417618

RESUMEN

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.


Asunto(s)
Antígenos/inmunología , Linaje de la Célula , Macrófagos/metabolismo , Enfermedades Metabólicas/prevención & control , Células Mieloides/metabolismo , Schistosoma mansoni/metabolismo , Esquistosomiasis mansoni/metabolismo , Animales , Reprogramación Celular , Dieta Alta en Grasa/efectos adversos , Femenino , Metabolismo de los Lípidos , Macrófagos/inmunología , Macrófagos/parasitología , Masculino , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/parasitología , Metaboloma , Ratones , Ratones Noqueados para ApoE , Células Mieloides/inmunología , Células Mieloides/parasitología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/parasitología
19.
FASEB J ; 35(1): e21205, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33337558

RESUMEN

CRISPR/Cas9-mediated genome editing shows cogent potential for the genetic modification of helminth parasites. We report successful gene knock-in (KI) into the genome of the egg of Schistosoma mansoni by combining CRISPR/Cas9 with single-stranded oligodeoxynucleotides (ssODNs). We edited the acetylcholinesterase (AChE) gene of S. mansoni targeting two guide RNAs (gRNAs), X5 and X7, located on exon 5 and exon 7 of Smp_154600, respectively. Eggs recovered from livers of experimentally infected mice were transfected by electroporation with a CRISPR/Cas9-vector encoding gRNA X5 or X7 combining with/ without a ssODN donor. Next generation sequencing analysis of reads of amplicon libraries spanning targeted regions revealed that the major modifications induced by CRISPR/Cas9 in the eggs were generated by homology directed repair (HDR). Furthermore, soluble egg antigen from AChE-edited eggs exhibited markedly reduced AChE activity, indicative that programed Cas9 cleavage mutated the AChE gene. Following injection of AChE-edited schistosome eggs into the tail veins of mice, an significantly enhanced Th2 response involving IL-4, -5, -10, and-13 was detected in lung cells and splenocytes in mice injected with X5-KI eggs in comparison to control mice injected with unmutated eggs. A Th2-predominant response, with increased levels of IL-4, -13, and GATA3, also was induced by X5 KI eggs in small intestine-draining mesenteric lymph node cells when the gene-edited eggs were introduced into the subserosa of the ileum of the mice. These findings confirmed the potential and the utility of CRISPR/Cas9-mediated genome editing for functional genomics in schistosomes.


Asunto(s)
Acetilcolinesterasa/metabolismo , Sistemas CRISPR-Cas , Edición Génica , Proteínas del Helminto/metabolismo , Schistosoma mansoni/enzimología , Esquistosomiasis mansoni/metabolismo , Acetilcolinesterasa/genética , Animales , Femenino , Proteínas del Helminto/genética , Ratones , Schistosoma mansoni/genética , Esquistosomiasis mansoni/genética
20.
J Comp Neurol ; 529(9): 2347-2361, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33368267

RESUMEN

Freshwater snails of the genus Biomphalaria serve as obligatory hosts for the digenetic trematode Schistosoma mansoni, the causative agent for the most widespread form of intestinal schistosomiasis. Within Biomphalaria, S. mansoni larvae multiply and transform into the cercariae form that can infect humans. Trematode development and proliferation is thought to be facilitated by modifications of host behavior and physiological processes, including a reduction of reproduction known as "parasitic castration." As neuropeptides participate in the control of reproduction across phylogeny, a neural transcriptomics approach was undertaken to identify peptides that could regulate Biomphalaria reproductive physiology. The present study identified a transcript in Biomphalaria alexandrina that encodes a peptide belonging to the gonadotropin-releasing hormone (GnRH) superfamily. The precursor and the predicted mature peptide, pQIHFTPDWGNN-NH2 (designated Biom-GnRH), share features with peptides identified in other molluscan species, including panpulmonates, opisthobranchs, and cephalopods. An antibody generated against Biom-GnRH labeled neurons in the cerebral, pedal, and visceral ganglia of Biomphalaria glabrata. GnRH-like immunoreactive fiber systems projected to all central ganglia. In the periphery, immunoreactive material was detected in the ovotestis, oviduct, albumen gland, and nidamental gland. As these structures serve crucial roles in the production, transport, nourishment, and encapsulation of eggs, disruption of the GnRH system of Biomphalaria could contribute to reduced reproductive activity in infected snails.


Asunto(s)
Biomphalaria/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Interacciones Huésped-Parásitos/fisiología , Schistosoma mansoni/metabolismo , Esquistosomiasis mansoni/metabolismo , Secuencia de Aminoácidos , Animales , Biomphalaria/química , Biomphalaria/genética , Hormona Liberadora de Gonadotropina/análisis , Hormona Liberadora de Gonadotropina/genética , Neuropéptidos , Schistosoma mansoni/genética , Esquistosomiasis mansoni/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...