Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
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
2.
PLoS Pathog ; 17(12): e1010064, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34969052

RESUMEN

Schistosomes are long lived, intravascular parasitic platyhelminths that infect >200 million people globally. The molecular mechanisms used by these blood flukes to dampen host immune responses are described in this review. Adult worms express a collection of host-interactive tegumental ectoenzymes that can cleave host signaling molecules such as the "alarmin" ATP (cleaved by SmATPDase1), the platelet activator ADP (SmATPDase1, SmNPP5), and can convert AMP into the anti-inflammatory mediator adenosine (SmAP). SmAP can additionally cleave the lipid immunomodulator sphingosine-1-phosphate and the proinflammatory anionic polymer, polyP. In addition, the worms release a barrage of proteins (e.g., SmCB1, SjHSP70, cyclophilin A) that can impinge on immune cell function. Parasite eggs also release their own immunoregulatory proteins (e.g., IPSE/α1, omega1, SmCKBP) as do invasive cercariae (e.g., Sm16, Sj16). Some schistosome glycans (e.g., LNFPIII, LNnT) and lipids (e.g., Lyso-PS, LPC), produced by several life stages, likewise affect immune cell responses. The parasites not only produce eicosanoids (e.g., PGE2, PGD2-that can be anti-inflammatory) but can also induce host cells to release these metabolites. Finally, the worms release extracellular vesicles (EVs) containing microRNAs, and these too have been shown to skew host cell metabolism. Thus, schistosomes employ an array of biomolecules-protein, lipid, glycan, nucleic acid, and more, to bend host biochemistry to their liking. Many of the listed molecules have been individually shown capable of inducing aspects of the polarized Th2 response seen following infection (with the generation of regulatory T cells (Tregs), regulatory B cells (Bregs) and anti-inflammatory, alternatively activated (M2) macrophages). Precisely how host cells integrate the impact of these myriad parasite products following natural infection is not known. Several of the schistosome immunomodulators described here are in development as novel therapeutics against autoimmune, inflammatory, and other, nonparasitic, diseases.


Asunto(s)
Proteínas del Helminto/inmunología , Factores Inmunológicos/inmunología , Schistosoma/inmunología , Esquistosomiasis/inmunología , Animales , Humanos
3.
Biochem J ; 479(11): 1165-1180, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35593185

RESUMEN

Infection with schistosomes (blood flukes) can result in the debilitating disease schistosomiasis. These parasites survive in their host for many years, and we hypothesize that proteins on their tegumental surface, interacting with the host microenvironment, facilitate longevity. One such ectoenzyme - the nucleotide pyrophosphatase/phosphodiesterase SmNPP5 can cleave ADP (to prevent platelet aggregation) and NAD (likely preventing Treg apoptosis). A second tegumental ectoenzyme, the glycohydrolase SmNACE, also catabolizes NAD. Here, we undertake a comparative biochemical characterization of these parasite ectoenzymes. Both are GPI-linked and exhibit different optimal pH ranges. While SmNPP5 requires divalent cations, SmNACE does not. The KM values of the two enzymes for NAD at physiological pH differ: SmNPP5, KM = 340 µM ± 44; SmNACE, KM = 49 µM ± 4. NAD cleavage by each enzyme yields different products. SmNPP5 cleaves NAD to form nicotinamide mononucleotide (NMN) and AMP, whereas SmNACE cleaves NAD to generate nicotinamide (NAM) and adenosine diphosphate ribose (ADPR). Each enzyme can process the other's reaction product. Thus, SmNACE cleaves NMN (to yield NAM and ribose phosphate) and SmNPP5 cleaves ADPR (yielding AMP and ribose phosphate). Metabolomic analysis of plasma containing adult worms supports the idea that these cleavage pathways are active in vivo. We hypothesize that a primary function of SmNPP5 is to cleave NAD to control host immune cell function and a primary function of SmNACE is to cleave NMN to generate the vital nutrient nicotinamide (vitamin B3) for convenient uptake by the worms. Chemical inhibition of one or both ectoenzymes could upset worm metabolism and control schistosome infection.


Asunto(s)
NAD , Schistosoma mansoni , Adenosina Difosfato Ribosa , Adenosina Monofosfato , Animales , NAD/metabolismo , Niacinamida
4.
Bioessays ; 41(12): e1900103, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31661165

RESUMEN

Schistosomes are intravascular parasitic helminths (blood flukes) that infect more than 200 million people globally. Proteomic analysis of the tegument (skin) of these worms has revealed the surprising presence of glycolytic enzymes on the parasite's external surface. Immunolocalization data as well as enzyme activity displayed by live worms confirm that functional glycolytic enzymes are indeed expressed at the host-parasite interface. Since these enzymes are traditionally considered to function intracellularly to drive glycolysis, in an extracellular location they are hypothesized to engage in novel "moonlighting" functions such as immune modulation and blood clot dissolution that promote parasite survival. For instance, several glycolytic enzymes can interact with plasminogen and promote its activation to the thrombolytic plasmin; some can inhibit complement function; some induce B cell proliferation or macrophage apoptosis. Several pathogenic bacteria and protists also express glycolytic enzymes externally, suggesting that moonlighting functions of extracellular glycolytic enzymes can contribute broadly to pathogen virulence. Also see the video abstract here https://youtu.be/njtWZ2y3k_I.


Asunto(s)
Proteínas del Helminto/metabolismo , Proteínas del Helminto/fisiología , Schistosoma/metabolismo , Schistosoma/patogenicidad , Animales , Glucólisis , Humanos , Proteómica/métodos , Esquistosomiasis/parasitología
5.
Int J Mol Sci ; 21(5)2020 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-32155992

RESUMEN

Schistosomiasis is a debilitating infection provoked by parasitic flatworms called schistosomes. The species Schistosoma mansoni is endemic in Africa, where it causes intestinal schistosomiasis. Recently, an α-carbonic anhydrase (CA, EC 4.2.1.1) was cloned and characterized from this organism and designated as SmCA. The protein is expressed in the tegument (skin) of S. mansoni at the host-parasite interface. Recombinant SmCA possesses high catalytic activity in the CO2 hydration reaction, similar to that of human CA isoform II with a kcat of 1.2 × 106 s-1 and a kcat/KM of 1.3 × 108 M-1·s-1. It has been found that schistosomes whose SmCA gene is suppressed using RNA interference are unable to establish a robust infection in mice, suggesting that the chemicals that inhibit SmCA function should have the same debilitating effect on the parasites. In this study, a collection of aromatic/heterocyclic sulfonamides were investigated as possible SmCA inhibitors. Several sulfonamides inhibited SmCA with medium to weak potency (KI values of 737.2 nM-9.25 µM), whereas some heterocyclic compounds inhibited the enzyme with KI values in the range of 124-325 nM. The α-CA from S. mansoni, SmCA, is proposed as a new anti-schistosomiasis drug target.


Asunto(s)
Antihelmínticos/farmacología , Inhibidores de Anhidrasa Carbónica/farmacología , Schistosoma mansoni/efectos de los fármacos , Esquistosomiasis/tratamiento farmacológico , Sulfonamidas/farmacología , Animales , Anhidrasas Carbónicas/genética , Anhidrasas Carbónicas/metabolismo , Humanos , Pruebas de Sensibilidad Parasitaria , Schistosoma mansoni/metabolismo , Relación Estructura-Actividad
6.
PLoS Pathog ; 11(12): e1005335, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26658895

RESUMEN

Schistosoma mansoni is a blood fluke parasite that causes schistosomiasis, a debilitating disease of global public health importance. These relatively large parasites are able to survive prolonged periods in the human vasculature without inducing stable blood clots around them. We show here that the intravascular life stages (schistosomula and adult males and females) can all promote significant plasminogen (PLMG) activation in the presence of tissue plasminogen activator (tPA). This results in the generation of the potent fibrinolytic agent plasmin which could degrade blood clots forming around the worms in vivo. We demonstrate that S. mansoni enolase (SmEno) is a host-interactive tegumental enzyme that, in recombinant form, can bind PLMG and promote its activation. Like classical members of the enolase protein family, SmEno can catalyze the interconversion of 2-phospho-D-glycerate (2-PGA) and phosphoenolpyruvate (PEP). The enzyme has maximal activity at pH 7.5, requires Mg2+ for optimal activity and can be inhibited by NaF but not mefloquin. Suppressing expression of the SmEno gene significantly diminishes enolase mRNA levels, protein levels and surface enzyme activity but, surprisingly, does not affect the ability of the worms to promote PLMG activation. Thus, while SmEno can enhance PLMG activation, our analysis suggests that it is not the only contributor to the parasite's ability to perform this function. We show that the worms possess several other PLMG-binding proteins in addition to SmEno and these may have a greater importance in schistosome-driven PLMG activation.


Asunto(s)
Proteínas del Helminto/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Plasminógeno/metabolismo , Schistosoma mansoni/enzimología , Esquistosomiasis mansoni/enzimología , Animales , Western Blotting , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección
7.
PLoS Pathog ; 10(8): e1004246, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25121497

RESUMEN

Schistosomes are parasitic flatworms that infect >200 million people worldwide, causing the chronic, debilitating disease schistosomiasis. Unusual among parasitic helminths, the long-lived adult worms, continuously bathed in blood, take up nutrients directly across the body surface and also by ingestion of blood into the gut. Recent proteomic analyses of the body surface revealed the presence of hydrolytic enzymes, solute, and ion transporters, thus emphasising its metabolic credentials. Furthermore, definition of the molecular mechanisms for the uptake of selected metabolites (glucose, certain amino acids, and water) establishes it as a vital site of nutrient acquisition. Nevertheless, the amount of blood ingested into the gut per day is considerable: for males ∼100 nl; for the more actively feeding females ∼900 nl, >4 times body volume. Ingested erythrocytes are lysed as they pass through the specialized esophagus, while leucocytes become tethered and disabled there. Proteomics and transcriptomics have revealed, in addition to gut proteases, an amino acid transporter in gut tissue and other hydrolases, ion, and lipid transporters in the lumen, implicating the gut as the site for acquisition of essential lipids and inorganic ions. The surface is the principal entry route for glucose, whereas the gut dominates amino acid acquisition, especially in females. Heme, a potentially toxic hemoglobin degradation product, accumulates in the gut and, since schistosomes lack an anus, must be expelled by the poorly understood process of regurgitation. Here we place the new observations on the proteome of body surface and gut, and the entry of different nutrient classes into schistosomes, into the context of older studies on worm composition and metabolism. We suggest that the balance between surface and gut in nutrition is determined by the constraints of solute diffusion imposed by differences in male and female worm morphology. Our conclusions have major implications for worm survival under immunological or pharmacological pressure.


Asunto(s)
Interacciones Huésped-Parásitos/fisiología , Schistosoma/fisiología , Animales , Femenino , Humanos , Masculino , Esquistosomiasis/fisiopatología
8.
J Thromb Thrombolysis ; 41(4): 671-7, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26573180

RESUMEN

Schistosomes are parasitic platyhelminths that currently infect over 200 million people and cause the chronic debilitating disease schistosomiasis. While these large intravascular parasites can disturb blood flow, surprisingly they do not appear to provoke thrombus formation around them in vivo. In order to determine if the worms can alter their local environment to impede coagulation, we incubated adult worms (50 pairs) in murine blood (500 µl) for 1 h at 37 °C and, using thromboelastography (TEG), we compared the coagulation profile of the blood with control blood that never contained worms. Substantial differences were apparent between the two profiles. Blood that had been exposed to schistosomes clotted more slowly and yielded relatively poor, though stable, thrombi; all TEG measures of blood coagulation (R, K, α-angle, MA, G and TMA) differed significantly between conditions. No fibrinolysis (as determined by LY30 and LY60 values) was detected in either case. The observed TEG profile suggests that the worms are acting as local anti-coagulants. Blood recovered from schistosome-infected mice, however, does not behave in this way. At an early time point post infection (4-weeks), the TEG profile of infected murine blood is essentially the same as that of control blood. However at a later time point (7-weeks) infected murine blood clots significantly faster than control blood but these clots also break down faster. The R, K, α-angle, and TMA measures of coagulation are all significantly different between the control versus infected mice as are the LY30 and LY60 values. This profile is indicative of a hypercoagulable state with fibrinolysis and is akin to that seen in human patients with advanced schistosomiasis.


Asunto(s)
Fibrinólisis , Schistosoma mansoni , Esquistosomiasis mansoni/sangre , Trombofilia , Trombosis , Animales , Femenino , Humanos , Ratones , Trombofilia/sangre , Trombofilia/parasitología , Trombosis/sangre , Trombosis/parasitología
9.
Purinergic Signal ; 10(4): 573-80, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24894599

RESUMEN

Schistosomes are parasitic worms that can live in the bloodstream of their vertebrate hosts for many years. It has been proposed that the worms impinge on host purinergic signalling by degrading proinflammatory molecules like ATP as well as prothrombotic mediators like ADP. This capability may help explain the apparent refractoriness of the worms to both immune elimination and thrombus formation. Three distinct ectoenzymes, expressed at the host-exposed surface of the worm's tegument, are proposed to be involved in the catabolism of ATP and ADP. These are alkaline phosphatase (SmAP), phosphodiesterase (SmNPP-5), and ATP diphosphohydrolase (SmATPDase1). It has recently been shown that only one of these enzymes-SmATPDase1-actually degrades exogenous ATP and ADP. However, a second ATP diphosphohydrolase homolog (SmATPDase2) is located in the tegument and has been reported to be released by the worms. It is possible that this enzyme too participates in the cleavage of exogenous nucleotide tri- and di-phosphates. To test this hypothesis, we employed RNA interference (RNAi) to suppress the expression of the schistosome SmATPDase1 and SmATPDase2 genes. We find that only SmATPDase1-suppressed parasites are significantly impaired in their ability to degrade exogenously added ATP or ADP. Suppression of SmATPDase2 does not appreciably affect the worms' ability to catabolize ATP or ADP. Furthermore, we detect no evidence for the secretion or release of an ATP-hydrolyzing activity by cultured parasites. The results confirm the role of tegumental SmATPDase1, but not SmADTPDase2, in the degradation of the exogenous proinflammatory and prothrombotic nucleotides ATP and ADP by live intravascular stages of the parasite.


Asunto(s)
Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Apirasa/metabolismo , Interacciones Huésped-Parásitos/fisiología , Schistosoma mansoni/enzimología , Esquistosomiasis mansoni/enzimología , Animales , Isoenzimas , Datos de Secuencia Molecular , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcriptoma , Transfección
10.
Heliyon ; 10(7): e28271, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38601580

RESUMEN

Schistosomes are intravascular parasitic worms infecting >200 million people globally. Here we examine how the worms acquire an essential nutrient - vitamin B2 (riboflavin). We demonstrate that all intravascular life stages (schistosomula, adult males and females) take up radiolabeled riboflavin. This process is impeded in the presence of excess unlabeled riboflavin and at 4 °C. We have identified a transporter homolog in worms designated SmaRT (Schistosoma mansoni riboflavin transporter) that localizes to the tegument and internal tissues of adults. CHO-S cells transfected with plasmid encoding SmaRT import significantly more radiolabeled riboflavin compared to controls. Uptake of radiolabel is impeded when SmaRT-expressing cells are incubated in an excess of unlabeled riboflavin but not by an excess of an irrelevant metabolite. Uptake is mediated in a sodium-independent manner and over a wide range of pH values (pH 5.5-9). This is the first identification of a bone fide riboflavin transporter in any platyhelminth.

11.
bioRxiv ; 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38558993

RESUMEN

Schistosomiasis is a parasitic disease caused by trematode worms 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. In this work we focus on the biochemistry of riboflavin and its metabolites in Schistosoma mansoni. We show that when schistosomes are incubated in murine plasma, levels of FAD decrease over time while the levels of FMN increase. We show that live schistosomes can cleave exogenous FAD to generate FMN and this ability is significantly blocked when expression of the surface ectoenzyme SmNPP5 is suppressed using RNAi. Recombinant SmNPP5 cleaves FAD with a Km of 178 ± 5.9 µM. The FAD-dependent enzyme IL-4I1 drives the oxidative deamination of phenylalanine to produce phenylpyruvate and H2O2 in the extracellular environment. Since schistosomes can be 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, suggesting that covalently bound FAD on IL-4I1 is inaccessible to SmNPP5. We also report here 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. Thus, 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, we hypothesize, the recently described schistosome riboflavin transporter SmaRT. In this work we also 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.

12.
J Parasitol ; 110(2): 96-105, 2024 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-38466806

RESUMEN

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.


Asunto(s)
Esquistosomiasis mansoni , Esquistosomiasis , Vacunas , Animales , Ratones , Schistosoma mansoni , Fosfoglicerato Mutasa , Esquistosomiasis mansoni/prevención & control , Esquistosomiasis mansoni/parasitología , Proteómica , Esquistosomiasis/prevención & control , Antígenos Helmínticos , Anticuerpos Antihelmínticos
13.
Front Immunol ; 14: 1056469, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36798133

RESUMEN

Schistosomes are long-lived parasitic worms that infect >200 million people globally. The intravascular life stages are known to display acetylcholinesterase (AChE) activity internally as well as, somewhat surprisingly, on external tegumental membranes. Originally it was hypothesized that a single gene (SmAChE1 in Schistosoma mansoni) encoded both forms of the enzyme. Here, we demonstrate that a second gene, designated "S. mansoni tegumental acetylcholinesterase, SmTAChE", is responsible for surface, non-neuronal AChE activity. The SmTAChE protein is GPI-anchored and contains all essential amino acids necessary for function. AChE surface activity is significantly diminished following SmTAChE gene suppression using RNAi, but not following SmAChE1 gene suppression. Suppressing SmTAChE significantly impairs the ability of parasites to establish infection in mice, showing that SmTAChE performs an essential function for the worms in vivo. Living S. haematobium and S. japonicum parasites also display strong surface AChE activity, and we have cloned SmTAChE homologs from these two species. This work helps to clarify longstanding confusion regarding schistosome AChEs and paves the way for novel therapeutics for schistosomiasis.


Asunto(s)
Parásitos , Esquistosomiasis , Animales , Ratones , Acetilcolinesterasa/genética , Acetilcolinesterasa/metabolismo , Parásitos/metabolismo , Schistosoma mansoni , Interferencia de ARN
14.
PLoS Pathog ; 6(6): e1000932, 2010 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-20532163

RESUMEN

Adult schistosomes live in the host's bloodstream where they import nutrients such as glucose across their body surface (the tegument). The parasite tegument is an unusual structure since it is enclosed not by the typical one but by two closely apposed lipid bilayers. Within the tegument two glucose importing proteins have been identified; these are schistosome glucose transporter (SGTP) 1 and 4. SGTP4 is present in the host interactive, apical tegumental membranes, while SGTP1 is found in the tegumental basal membrane (as well as in internal tissues). The SGTPs act by facilitated diffusion. To examine the importance of these proteins for the parasites, RNAi was employed to knock down expression of both SGTP genes in the schistosomula and adult worm life stages. Both qRT-PCR and western blotting analysis confirmed successful gene suppression. It was found that SGTP1 or SGTP4-suppressed parasites exhibit an impaired ability to import glucose compared to control worms. In addition, parasites with both SGTP1 and SGTP4 simultaneously suppressed showed a further reduction in capacity to import glucose compared to parasites with a single suppressed SGTP gene. Despite this debility, all suppressed parasites exhibited no phenotypic distinction compared to controls when cultured in rich medium. Following prolonged incubation in glucose-depleted medium however, significantly fewer SGTP-suppressed parasites survived. Finally, SGTP-suppressed parasites showed decreased viability in vivo following infection of experimental animals. These findings provide direct evidence for the importance of SGTP1 and SGTP4 for schistosomes in importing exogenous glucose and show that these proteins are important for normal parasite development in the mammalian host.


Asunto(s)
Conducta Alimentaria , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Ratones Endogámicos BALB C/parasitología , Schistosoma mansoni/crecimiento & desarrollo , Schistosoma mansoni/metabolismo , Animales , Western Blotting , Supervivencia Celular , Transportador de Glucosa de Tipo 1/antagonistas & inhibidores , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 4/antagonistas & inhibidores , Transportador de Glucosa de Tipo 4/genética , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Caracoles/parasitología , Tasa de Supervivencia
15.
Acta Trop ; 236: 106676, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36113567

RESUMEN

Schistosomes are intravascular parasitic platyhelminths (blood flukes) that infect over 200 million people globally. Biomolecules secreted by the worms likely contribute to their ability to survive in the bloodstreams of immunocompetent hosts for many years. Here we review what is known about the protein composition of material released by the worms. Prominent among cercarial excretions/secretions (ES) is a ∼ 30 kDa serine protease called cercarial elastase (SmCE in Schistosoma mansoni), likely important in host invasion. Also prominent is a 117 amino acid non-glycosylated polypeptide (Sm16) that can impact several host cell-types to impinge on immunological outcomes. Similarly, components of the egg secretome (notably the 134 amino acid homodimeric glycoprotein "IL-4 inducing principle of schistosome eggs", IPSE, and the 225-amino acid monomeric T2 ribonuclease - omega-1) are capable of driving Th2-biased immune responses. A ∼36kDa chemokine binding glycoprotein SmCKBP, secreted by eggs, can negate the impact of several cytokines and can impede neutrophil migration. Of special interest is a disparate collection of classically cytosolic proteins that are surprisingly often identified in schistosome ES across life stages. These proteins, perhaps released as components of extracellular vesicles (EVs), include glycolytic enzymes, redox proteins, proteases and protease inhibitors, heat shock proteins, proteins involved in translation/turnover, histones, and others. Some such proteins may display "moonlighting" functions and, for example, impede blood clot formation around the worms. More prosaically, since several are particularly abundant soluble proteins, their appearance in the ES fraction may be indicative of worm damage ex vivo leading to protein leakage. Some bioactive schistosome ES proteins are in development as novel therapeutics against autoimmune, inflammatory, and other, non-parasitic, diseases.


Asunto(s)
Proteínas del Helminto , Histonas , Aminoácidos/metabolismo , Animales , Quimiocinas/metabolismo , Glicoproteínas , Proteínas de Choque Térmico/metabolismo , Proteínas del Helminto/metabolismo , Histonas/metabolismo , Interleucina-4 , Elastasa Pancreática/metabolismo , Inhibidores de Proteasas , Ribonucleasas/metabolismo , Schistosoma mansoni , Secretoma , Serina Proteasas/metabolismo
16.
Trends Parasitol ; 38(12): 1080-1088, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36182536

RESUMEN

Intravascular schistosomes may control immune and hemostatic responses by regulating the nature and amount of selected host purinergic signaling molecules - such as adenosine triphosphate (ATP), adenosine diphosphate (ADP), and nicotinamide adenine dinucleotide (NAD) - surrounding them. Such metabolites are collectively known as the worm's 'purinergic halo'. Host-interactive, membrane-bound, tegumental ectonucleotidases, notably SmATPDase1, SmNPP5, SmAP and SmNACE, can degrade proinflammatory, prothrombotic and immunomodulatory purinergic metabolites like those listed. A common catabolic product is the anti-inflammatory metabolite adenosine that can additionally be taken in by the worms as food. We envision the tegumental ectonucleotidases as having a twofold role at the worm surface: first, they degrade potentially harmful host signaling molecules, and second, they generate vital nutrients around the worms from where these can be conveniently imported.


Asunto(s)
Schistosoma mansoni , Animales
17.
Parasite ; 29: 41, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36083036

RESUMEN

Schistosomiasis is a debilitating parasitic disease caused by intravascular flatworms called schistosomes (blood flukes) that affects >200 million people worldwide. Proteomic analysis has revealed the surprising presence of classical glycolytic enzymes - typically cytosolic proteins - located on the extracellular surface of the parasite tegument (skin). Immunolocalization experiments show that phosphoglycerate mutase (PGM) is widely expressed in parasite tissues and is highly expressed in the tegument. We demonstrate that live Schistosoma mansoni parasites express enzymatically active PGM on their tegumental surface. Suppression of PGM using RNA interference (RNAi) diminishes S. mansoni PGM (SmPGM) gene expression, protein levels, and surface enzyme activity. Sequence comparisons place SmPGM in the cofactor (2,3-bisphosphoglycerate)-dependent PGM (dPGM) family. We have produced recombinant SmPGM (rSmPGM) in an enzymatically active form in Escherichia coli. The Michaelis-Menten constant (Km) of rSmPGM for its glycolytic substrate (3-phosphoglycerate) is 0.85 mM ± 0.02. rSmPGM activity is inhibited by the dPGM-specific inhibitor vanadate. Here, we show that rSmPGM not only binds to plasminogen but also promotes its conversion to an active form (plasmin) in vitro. This supports the hypothesis that host-interactive tegumental proteins (such as SmPGM), by enhancing plasmin formation, may help degrade blood clots around the worms in the vascular microenvironment and thus promote parasite survival in vivo.


Title: La phosphoglycérate mutase de Schistosoma mansoni ­ une ectoenzyme glycolytique avec un potentiel thrombolytique. Abstract: La schistosomiase est une maladie parasitaire débilitante causée par des vers plats intravasculaires appelés schistosomes qui affecte plus de 200 millions de personnes dans le monde. L'analyse protéomique a révélé la présence surprenante d'enzymes glycolytiques classiques ­ typiquement des protéines cytosoliques ­ situées sur la surface extracellulaire du tégument du parasite. Des expériences d'immunolocalisation montrent que la phosphoglycérate mutase (PGM) est largement exprimée dans les tissus parasitaires et fortement exprimée dans le tégument. Nous démontrons que les parasites Schistosoma mansoni vivants expriment une PGM enzymatiquement active sur leur surface tégumentaire. La suppression de la PGM à l'aide de l'interférence ARN (ARNi) diminue l'expression du gène PGM de S. mansoni (SmPGM), les niveaux de protéines et l'activité enzymatique de surface. Les comparaisons de séquences placent la SmPGM dans la famille des PGM dépendantes du cofacteur (2,3-bisphosphoglycérate) (dPGM). Nous avons produit de la SmPGM recombinante (rSmPGM) sous une forme enzymatiquement active dans Escherichia coli. La constante de Michaelis-Menten (Km) de rSmPGM pour son substrat glycolytique (3-phosphoglycérate) est de 0,85 mM ± 0,02. L'activité de la rSmPGM est inhibée par le vanadate, un inhibiteur spécifique des dPGM. Ici, nous montrons que rSmPGM non seulement se lie au plasminogène mais favorise également sa conversion en une forme active (plasmine) in vitro. Cela soutient l'hypothèse selon laquelle les protéines tégumentaires interactives avec l'hôte (telles que SmPGM), en améliorant la formation de plasmine, peuvent aider à dégrader les caillots sanguins autour des vers dans le microenvironnement vasculaire et ainsi favoriser la survie du parasite in vivo.


Asunto(s)
Schistosoma mansoni/enzimología , Esquistosomiasis , Animales , Fibrinolisina/metabolismo , Interacciones Huésped-Parásitos , Humanos , Fosfoglicerato Mutasa/genética , Fosfoglicerato Mutasa/metabolismo , Proteómica , Esquistosomiasis/parasitología
18.
Pathogens ; 11(2)2022 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-35215099

RESUMEN

Schistosomes (blood flukes) can survive in the bloodstream of their hosts for many years. We hypothesize that proteins on their host-interactive surface impinge on host biochemistry to help ensure their long-term survival. Here, we focus on a surface ectoenzyme of Schistosoma mansoni, designated SmNPP5. This ~53 kDa glycoprotein is a nucleotide pyrophosphatase/phosphodiesterase that has been previously shown to: (1) cleave adenosine diphosphate (ADP) and block platelet aggregation; and (2) cleave nicotinamide adenine dinucleotide (NAD) and block NAD-induced T cell apoptosis in vitro. T cell apoptosis can additionally be driven by extracellular adenosine triphosphate (ATP). In this work, we show that adult S. mansoni parasites can inhibit this process. Further, we demonstrate that recombinant SmNPP5 alone can both cleave ATP and impede ATP-induced T cell killing. As immunomodulatory regulatory T cells (Tregs) are especially prone to the induction of these apoptotic pathways, we hypothesize that the schistosome cleavage of both NAD and ATP promotes Treg survival and this helps to create a less immunologically hostile environment for the worms in vivo.

19.
Infect Immun ; 79(10): 4276-84, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21825060

RESUMEN

The intravascular trematode Schistosoma mansoni is a causative agent of schistosomiasis, a disease that constitutes a major health problem globally. In this study we cloned and characterized the schistosome tegumental phosphodiesterase SmNPP-5 and evaluated its role in parasite virulence. SmNPP-5 is a 52.5-kDa protein whose gene is rapidly turned on in the intravascular parasitic life stages, following invasion of the definitive host. Highest expression is found in mated adult males. As revealed by immunofluorescence analysis, SmNPP-5 protein is found prominently in the dorsal surface of the tegument of males. Localization by immuno-electron microscopy illustrates a unique pattern of immunogold-labeled SmNPP-5 within the tegument; some immunogold particles are scattered throughout the tissue, but many are clustered in tight arrays. To determine the importance of the protein for the parasites, RNA interference (RNAi) was employed to knock down expression of the SmNPP-5-encoding gene in schistosomula and adult worms. Both quantitative real-time PCR (qRT-PCR) and Western blotting confirmed successful and robust gene suppression. In addition, the suppression and the ectolocalization of this enzyme in live parasites were evident because of a significantly impaired ability of the suppressed parasites to hydrolyze exogenously added phosphodiesterase substrate p-nitrophenyl 5'-dTMP (p-Nph-5'-TMP). The effects of suppressing expression of the SmNPP-5 gene in vivo were tested by injecting parasites into mice. It was found that, unlike controls, parasites whose SmNPP-5 gene was demonstrably suppressed at the time of host infection were greatly impaired in their ability to establish infection. These results demonstrate that SmNPP-5 is a virulence factor for schistosomes.


Asunto(s)
Proteínas del Helminto/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Schistosoma mansoni/enzimología , Schistosoma mansoni/patogenicidad , Esquistosomiasis mansoni/parasitología , Factores de Virulencia/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , Femenino , Proteínas del Helminto/química , Proteínas del Helminto/genética , Masculino , Ratones , Microscopía Inmunoelectrónica , Datos de Secuencia Molecular , Hidrolasas Diéster Fosfóricas/química , Hidrolasas Diéster Fosfóricas/genética , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Schistosoma mansoni/crecimiento & desarrollo , Análisis de Secuencia de ADN , Virulencia , Factores de Virulencia/química , Factores de Virulencia/genética
20.
Front Mol Biosci ; 8: 719678, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34458323

RESUMEN

Fructose 1,6-bisphosphate aldolase is a ubiquitous cytosolic enzyme that catalyzes the fourth step of glycolysis. Aldolases are classified into three groups: Class-I, Class-IA, and Class-II; all classes share similar structural features but low amino acid identity. Apart from their conserved role in carbohydrate metabolism, aldolases have been reported to perform numerous non-enzymatic functions. Here we review the myriad "moonlighting" functions of this classical enzyme, many of which are centered on its ability to bind to an array of partner proteins that impact cellular scaffolding, signaling, transcription, and motility. In addition to the cytosolic location, aldolase has been found the extracellular surface of several pathogenic bacteria, fungi, protozoans, and metazoans. In the extracellular space, the enzyme has been reported to perform virulence-enhancing moonlighting functions e.g., plasminogen binding, host cell adhesion, and immunomodulation. Aldolase's importance has made it both a drug target and vaccine candidate. In this review, we note the several inhibitors that have been synthesized with high specificity for the aldolases of pathogens and cancer cells and have been shown to inhibit classical enzyme activity and moonlighting functions. We also review the many trials in which recombinant aldolases have been used as vaccine targets against a wide variety of pathogenic organisms including bacteria, fungi, and metazoan parasites. Most of such trials generated significant protection from challenge infection, correlated with antigen-specific cellular and humoral immune responses. We argue that refinement of aldolase antigen preparations and expansion of immunization trials should be encouraged to promote the advancement of promising, protective aldolase vaccines.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA