Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.749
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nat Immunol ; 18(6): 642-653, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28436955

RESUMEN

It remains unclear whether activated inflammatory macrophages can adopt features of tissue-resident macrophages, or what mechanisms might mediate such a phenotypic conversion. Here we show that vitamin A is required for the phenotypic conversion of interleukin 4 (IL-4)-activated monocyte-derived F4/80intCD206+PD-L2+MHCII+ macrophages into macrophages with a tissue-resident F4/80hiCD206-PD-L2-MHCII-UCP1+ phenotype in the peritoneal cavity of mice and during the formation of liver granulomas in mice infected with Schistosoma mansoni. The phenotypic conversion of F4/80intCD206+ macrophages into F4/80hiCD206- macrophages was associated with almost complete remodeling of the chromatin landscape, as well as alteration of the transcriptional profiles. Vitamin A-deficient mice infected with S. mansoni had disrupted liver granuloma architecture and increased mortality, which indicates that failure to convert macrophages from the F4/80intCD206+ phenotype to F4/80hiCD206- may lead to dysregulated inflammation during helminth infection.


Asunto(s)
Granuloma/inmunología , Hígado/inmunología , Macrófagos/inmunología , Esquistosomiasis mansoni/inmunología , Deficiencia de Vitamina A/inmunología , Animales , Antígenos de Diferenciación/metabolismo , Citometría de Flujo , Antígenos de Histocompatibilidad Clase II/metabolismo , Interleucina-4/inmunología , Lectinas Tipo C/metabolismo , Hígado/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Cavidad Peritoneal/citología , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Superficie Celular/metabolismo , Schistosoma mansoni , Esquistosomiasis mansoni/patología , Tretinoina/farmacología , Proteína Desacopladora 1/metabolismo , Vitaminas/farmacología
2.
Proc Natl Acad Sci U S A ; 121(2): e2315463120, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38181058

RESUMEN

Schistosomiasis is a neglected tropical disease affecting over 150 million people. Hotspots of Schistosoma transmission-communities where infection prevalence does not decline adequately with mass drug administration-present a key challenge in eliminating schistosomiasis. Current approaches to identify hotspots require evaluation 2-5 y after a baseline survey and subsequent mass drug administration. Here, we develop statistical models to predict hotspots at baseline prior to treatment comparing three common hotspot definitions, using epidemiologic, survey-based, and remote sensing data. In a reanalysis of randomized trials in 589 communities in five endemic countries, a regression model predicts whether Schistosoma mansoni infection prevalence will exceed the WHO threshold of 10% in year 5 ("prevalence hotspot") with 86% sensitivity, 74% specificity, and 93% negative predictive value (NPV; assuming 30% hotspot prevalence), and a regression model for Schistosoma haematobium achieves 90% sensitivity, 90% specificity, and 96% NPV. A random forest model predicts whether S. mansoni moderate and heavy infection prevalence will exceed a public health goal of 1% in year 5 ("intensity hotspot") with 92% sensitivity, 79% specificity, and 96% NPV, and a boosted trees model for S. haematobium achieves 77% sensitivity, 95% specificity, and 91% NPV. Baseline prevalence is a top predictor in all models. Prediction is less accurate in countries not represented in training data and for a third hotspot definition based on relative prevalence reduction over time ("persistent hotspot"). These models may be a tool to prioritize high-risk communities for more frequent surveillance or intervention against schistosomiasis, but prediction of hotspots remains a challenge.


Asunto(s)
Esquistosomiasis mansoni , Esquistosomiasis , Humanos , Animales , Administración Masiva de Medicamentos , Esquistosomiasis/tratamiento farmacológico , Esquistosomiasis/epidemiología , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/epidemiología , Schistosoma haematobium , Modelos Estadísticos
3.
PLoS Pathog ; 20(5): e1012268, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38814989

RESUMEN

The eggs of the blood fluke Schistosoma mansoni are the main cause of the clinical manifestations of chronic schistosomiasis. After laying, the egg "winners" attach to the endothelium of the mesenteric vein and, after a period of development, induce the growth of a small granuloma, which facilitates their passage to the intestinal lumen. Egg "losers" carried by the bloodstream to non-specific tissues also undergo full development and induce large granuloma formation, but their life ends there. Although these trapped eggs represent a dead end in the parasite life cycle, the vast majority of studies attempting to describe the biology of the S. mansoni eggs have studied these liver-trapped "losers" instead of migrating intestinal "winners". This raises the fundamental question of how these eggs differ. With robust comparative transcriptomic analysis performed on S. mansoni eggs isolated 7 weeks post infection, we show that gene expression is critically dependent on tissue localization, both in the early and late stages of development. While mitochondrial genes and venom allergen-like proteins are significantly upregulated in mature intestinal eggs, well-described egg immunomodulators IPSE/alpha-1 and omega-1, together with micro-exon genes, are predominantly expressed in liver eggs. In addition, several proteases and protease inhibitors previously implicated in egg-host interactions display clear tissue-specific gene expression patterns. These major differences in gene expression could be then reflected in the observed different ability of liver and intestinal soluble egg antigens to elicit host immune responses and in the shorter viability of miracidia hatched from liver eggs. Our comparative analysis provides a new perspective on the biology of parasite's eggs in the context of their development and tissue localization. These findings could contribute to a broader and more accurate understanding of parasite eggs interactions with the host, which have historically been often restricted to liver eggs and sometimes inaccurately generalized.


Asunto(s)
Hígado , Schistosoma mansoni , Esquistosomiasis mansoni , Animales , Schistosoma mansoni/inmunología , Schistosoma mansoni/genética , Hígado/parasitología , Hígado/inmunología , Hígado/metabolismo , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/parasitología , Ratones , Óvulo/metabolismo , Óvulo/inmunología , Intestinos/parasitología , Intestinos/inmunología , Antígenos Helmínticos/inmunología , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Proteínas del Helminto/inmunología , Femenino , Proteínas del Huevo
4.
Proc Natl Acad Sci U S A ; 120(8): e2211703120, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36780522

RESUMEN

The immune system is increasingly recognized as an important regulator of tissue repair. We developed a regenerative immunotherapy from the helminth Schistosoma mansoni soluble egg antigen (SEA) to stimulate production of interleukin (IL)-4 and other type 2-associated cytokines without negative infection-related sequelae. The regenerative SEA (rSEA) applied to a murine muscle injury induced accumulation of IL-4-expressing T helper cells, eosinophils, and regulatory T cells and decreased expression of IL-17A in gamma delta (γδ) T cells, resulting in improved repair and decreased fibrosis. Encapsulation and controlled release of rSEA in a hydrogel further enhanced type 2 immunity and larger volumes of tissue repair. The broad regenerative capacity of rSEA was validated in articular joint and corneal injury models. These results introduce a regenerative immunotherapy approach using natural helminth derivatives.


Asunto(s)
Esquistosomiasis mansoni , Animales , Ratones , Esquistosomiasis mansoni/terapia , Citocinas/metabolismo , Schistosoma mansoni , Linfocitos T Colaboradores-Inductores , Antígenos Helmínticos , Inmunoterapia
5.
J Biol Chem ; 300(1): 105528, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38043794

RESUMEN

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.


Asunto(s)
Antihelmínticos , Clonazepam , Esquistosomiasis mansoni , Canales Catiónicos TRPM , Animales , Humanos , Antihelmínticos/farmacología , Benzodiazepinas/farmacología , Benzodiazepinonas/farmacología , Clonazepam/análogos & derivados , Clonazepam/farmacología , Praziquantel/farmacología , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/metabolismo , Esquistosomiasis mansoni/tratamiento farmacológico , Canales Catiónicos TRPM/agonistas
6.
PLoS Pathog ; 19(5): e1011369, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37146077

RESUMEN

The trematode parasite Schistosoma mansoni causes schistosomiasis, which affects over 200 million people worldwide. Schistosomes are dioecious, with egg laying depending on the females' obligatory pairing with males. Long non-coding RNAs (lncRNAs) are transcripts longer than 200 nucleotides with low or no protein-coding potential that have been involved in other species with reproduction, stem cell maintenance, and drug resistance. In S. mansoni, we recently showed that the knockdown of one lncRNA affects the pairing status of these parasites. Here, we re-analyzed public RNA-Seq data from paired and unpaired adult male and female worms and their gonads, obtained from mixed-sex or single-sex cercariae infections, and found thousands of differentially expressed pairing-dependent lncRNAs among the 23 biological samples that were compared. The expression levels of selected lncRNAs were validated by RT-qPCR using an in vitro unpairing model. In addition, the in vitro silencing of three selected lncRNAs showed that knockdown of these pairing-dependent lncRNAs reduced cell proliferation in adult worms and their gonads, and are essential for female vitellaria maintenance, reproduction, and/or egg development. Remarkably, in vivo silencing of each of the three selected lncRNAs significantly reduced worm burden in infected mice by 26 to 35%. Whole mount in situ hybridization experiments showed that these pairing-dependent lncRNAs are expressed in reproductive tissues. These results show that lncRNAs are key components intervening in S. mansoni adult worm homeostasis, which affects pairing status and survival in the mammalian host, thus presenting great potential as new therapeutic target candidates.


Asunto(s)
Parásitos , ARN Largo no Codificante , Esquistosomiasis mansoni , Masculino , Femenino , Animales , Ratones , Schistosoma mansoni/genética , ARN Largo no Codificante/genética , Fertilidad/genética , Reproducción , Parásitos/genética , Esquistosomiasis mansoni/parasitología , Mamíferos
7.
PLoS Pathog ; 19(2): e1010884, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36730464

RESUMEN

Schistosoma mansoni employs immune evasion and immunosuppression to overcome immune responses mounted by its snail and human hosts. Myriad immunomodulating factors underlie this process, some of which are proteases. Here, we demonstrate that one protease, an invadolysin we have termed SmCI-1, is released from the acetabular glands of S. mansoni cercaria and is involved in creating an immunological milieu favorable for survival of the parasite. The presence of SmCI-1 in the cercarial stage of S. mansoni is released during transformation into the schistosomula. SmCI-1 functions as a metalloprotease with the capacity to cleave collagen type IV, gelatin and fibrinogen. Additionally, complement component C3b is cleaved by this protease, resulting in inhibition of the classical and alternative complement pathways. Using SmCI-1 knockdown cercariae, we demonstrate that SmCI-1 protects schistosomula from complement-mediated lysis in human plasma. We also assess the effect of SmCI-1 on cytokine release from human peripheral blood mononuclear cells, providing compelling evidence that SmCI-1 promotes an anti-inflammatory microenvironment by enhancing production of IL-10 and suppressing the production of inflammatory cytokines like IL-1B and IL-12p70 and those involved in eosinophil recruitment and activation, like Eotaxin-1 and IL-5. Finally, we utilize the SmCI-1 knockdown cercaria in a mouse model of infection, revealing a role for SmCI-1 in S. mansoni survival.


Asunto(s)
Schistosoma mansoni , Esquistosomiasis mansoni , Animales , Ratones , Humanos , Leucocitos Mononucleares , Cercarias , Proteínas del Sistema Complemento , Metaloproteasas , Inmunidad
8.
PLoS Pathog ; 19(7): e1011018, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37428793

RESUMEN

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


Asunto(s)
Antihelmínticos , Esquistosomiasis mansoni , Esquistosomiasis , Animales , Humanos , Praziquantel/farmacología , Praziquantel/química , Oxamniquina/farmacología , Esquistosomiasis/tratamiento farmacológico , Esquistosomiasis/parasitología , Schistosoma mansoni , Terapia Combinada , Enfermedades Desatendidas/tratamiento farmacológico , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/parasitología
9.
PLoS Pathog ; 19(5): e1011037, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37228019

RESUMEN

Schistosoma haematobium is the most prevalent of the human-infecting schistosome species, causing significant morbidity in endemically exposed populations. Despite this, it has been relatively understudied compared to its fellow species, S. mansoni. Here we provide the first comprehensive characterization of the S. haematobium Tegument Allergen-Like protein family, a key protein family directly linked to protective immunity in S. mansoni infection. Comparable with observations for S. mansoni, parasite phylogenetic analysis and relative gene expression combined with host serological analysis support a cross-reactive relationship between S. haematobium TAL proteins, exposed to the host immune system as adult worms die, and closely related proteins, exposed during penetration by the infecting cercarial and early schistosomulae stages. Specifically, our results strengthen the evidence for host immunity driven by cross-reactivity between family members TAL3 and TAL5, establishing it for the first time for S. haematobium infection. Furthermore, we build upon this relationship to include the involvement of an additional member of the TAL protein family, TAL11 for both schistosome species. Finally, we show a close association between experience of infection and intensity of transmission and the development of protective IgE responses to these antigens, thus improving our knowledge of the mechanisms by which protective host immune responses develop. This knowledge will be critical in understanding how control efforts such as mass drug administration campaigns influence the development of host immunity and subsequent patterns of infection and disease within endemic populations.


Asunto(s)
Schistosoma haematobium , Esquistosomiasis mansoni , Adulto , Animales , Humanos , Schistosoma haematobium/genética , Schistosoma mansoni/genética , Alérgenos , Filogenia , Estadios del Ciclo de Vida , Inmunoglobulina E
10.
Immunity ; 45(1): 145-58, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27421703

RESUMEN

Fibroproliferative diseases are driven by dysregulated tissue repair responses and are a major cause of morbidity and mortality because they affect nearly every organ system. Type 2 cytokine responses are critically involved in tissue repair; however, the mechanisms that regulate beneficial regeneration versus pathological fibrosis are not well understood. Here, we have shown that the type 2 effector cytokine interleukin-13 simultaneously, yet independently, directed hepatic fibrosis and the compensatory proliferation of hepatocytes and biliary cells in progressive models of liver disease induced by interleukin-13 overexpression or after infection with Schistosoma mansoni. Using transgenic mice with interleukin-13 signaling genetically disrupted in hepatocytes, cholangiocytes, or resident tissue fibroblasts, we have revealed direct and distinct roles for interleukin-13 in fibrosis, steatosis, cholestasis, and ductular reaction. Together, these studies show that these mechanisms are simultaneously controlled but distinctly regulated by interleukin-13 signaling. Thus, it may be possible to promote interleukin-13-dependent hepatobiliary expansion without generating pathological fibrosis. VIDEO ABSTRACT.


Asunto(s)
Hígado Graso/inmunología , Fibroblastos/inmunología , Interleucina-13/metabolismo , Cirrosis Hepática Biliar/inmunología , Hígado/patología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Animales , Ácidos y Sales Biliares/biosíntesis , Proliferación Celular , Células Cultivadas , Fibrosis , Humanos , Interleucina-13/genética , Interleucina-13/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Células Th2/inmunología
11.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35121663

RESUMEN

Predicting and disrupting transmission of human parasites from wildlife hosts or vectors remains challenging because ecological interactions can influence their epidemiological traits. Human schistosomes, parasitic flatworms that cycle between freshwater snails and humans, typify this challenge. Human exposure risk, given water contact, is driven by the production of free-living cercariae by snail populations. Conventional epidemiological models and management focus on the density of infected snails under the assumption that all snails are equally infectious. However, individual-level experiments contradict this assumption, showing increased production of schistosome cercariae with greater access to food resources. We built bioenergetics theory to predict how resource competition among snails drives the temporal dynamics of transmission potential to humans and tested these predictions with experimental epidemics and demonstrated consistency with field observations. This resource-explicit approach predicted an intense pulse of transmission potential when snail populations grow from low densities, i.e., when per capita access to resources is greatest, due to the resource-dependence of cercarial production. The experiment confirmed this prediction, identifying a strong effect of infected host size and the biomass of competitors on per capita cercarial production. A field survey of 109 waterbodies also found that per capita cercarial production decreased as competitor biomass increased. Further quantification of snail densities, sizes, cercarial production, and resources in diverse transmission sites is needed to assess the epidemiological importance of resource competition and support snail-based disruption of schistosome transmission. More broadly, this work illustrates how resource competition can sever the correspondence between infectious host density and transmission potential.


Asunto(s)
Biomphalaria/parasitología , Interacciones Huésped-Parásitos/fisiología , Schistosoma mansoni/patogenicidad , Esquistosomiasis mansoni/parasitología , Caracoles/parasitología , Animales , Humanos
12.
Clin Infect Dis ; 78(Supplement_2): S126-S130, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38662698

RESUMEN

BACKGROUND: The 2030 target for schistosomiasis is elimination as a public health problem (EPHP), achieved when the prevalence of heavy-intensity infection among school-aged children (SAC) reduces to <1%. To achieve this, the new World Health Organization guidelines recommend a broader target of population to include pre-SAC and adults. However, the probability of achieving EPHP should be expected to depend on patterns in repeated uptake of mass drug administration by individuals. METHODS: We employed 2 individual-based stochastic models to evaluate the impact of school-based and community-wide treatment and calculated the number of rounds required to achieve EPHP for Schistosoma mansoni by considering various levels of the population never treated (NT). We also considered 2 age-intensity profiles, corresponding to a low and high burden of infection in adults. RESULTS: The number of rounds needed to achieve this target depends on the baseline prevalence and the coverage used. For low- and moderate-transmission areas, EPHP can be achieved within 7 years if NT ≤10% and NT <5%, respectively. In high-transmission areas, community-wide treatment with NT <1% is required to achieve EPHP. CONCLUSIONS: The higher the intensity of transmission, and the lower the treatment coverage, the lower the acceptable value of NT becomes. Using more efficacious treatment regimens would permit NT values to be marginally higher. A balance between target treatment coverage and NT values may be an adequate treatment strategy depending on the epidemiological setting, but striving to increase coverage and/or minimize NT can shorten program duration.


Asunto(s)
Erradicación de la Enfermedad , Schistosoma mansoni , Esquistosomiasis mansoni , Humanos , Esquistosomiasis mansoni/epidemiología , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/prevención & control , Niño , Animales , Adolescente , Schistosoma mansoni/efectos de los fármacos , Adulto , Prevalencia , Administración Masiva de Medicamentos , Salud Pública , Adulto Joven , Preescolar , Antihelmínticos/uso terapéutico , Antihelmínticos/administración & dosificación , Masculino , Femenino , Persona de Mediana Edad
13.
Clin Infect Dis ; 78(1): 90-93, 2024 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-37585653

RESUMEN

In a cross-sectional analysis of 354 Ugandan children (age 12-48 months) infected with Schistosoma mansoni, we assessed relationships between infection intensity and nutritional morbidities. Higher intensity was associated with an increased risk for anemia (RR = 1.05, 95% confidence interval [CI] 1.01-1.10) yet not associated with risk for underweight, stunting, or wasting.


Asunto(s)
Anemia , Esquistosomiasis mansoni , Niño , Animales , Humanos , Preescolar , Lactante , Esquistosomiasis mansoni/complicaciones , Esquistosomiasis mansoni/epidemiología , Uganda/epidemiología , Estado Nutricional , Estudios Transversales , Prevalencia , Schistosoma mansoni , Anemia/epidemiología , Anemia/etiología
14.
Clin Infect Dis ; 78(Supplement_2): S153-S159, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38662699

RESUMEN

BACKGROUND: Control of schistosomiasis (SCH) relies on the regular distribution of preventive chemotherapy (PC) over many years. For the sake of sustainable SCH control, a decision must be made at some stage to scale down or stop PC. These "stopping decisions" are based on population surveys that assess whether infection levels are sufficiently low. However, the limited sensitivity of the currently used diagnostic (Kato-Katz [KK]) to detect low-intensity infections is a concern. Therefore, the use of new, more sensitive, molecular diagnostics has been proposed. METHODS: Through statistical analysis of Schistosoma mansoni egg counts collected from Burundi and a simulation study using an established transmission model for schistosomiasis, we investigated the extent to which more sensitive diagnostics can improve decision making regarding stopping or continuing PC for the control of S. mansoni. RESULTS: We found that KK-based strategies perform reasonably well for determining when to stop PC at a local scale. Use of more sensitive diagnostics leads to a marginally improved health impact (person-years lived with heavy infection) and comes at a cost of continuing PC for longer (up to around 3 years), unless the decision threshold for stopping PC is adapted upward. However, if this threshold is set too high, PC may be stopped prematurely, resulting in a rebound of infection levels and disease burden (+45% person-years of heavy infection). CONCLUSIONS: We conclude that the potential value of more sensitive diagnostics lies more in the reduction of survey-related costs than in the direct health impact of improved parasite control.


Asunto(s)
Análisis Costo-Beneficio , Recuento de Huevos de Parásitos , Schistosoma mansoni , Esquistosomiasis mansoni , Humanos , Animales , Schistosoma mansoni/aislamiento & purificación , Esquistosomiasis mansoni/diagnóstico , Esquistosomiasis mansoni/prevención & control , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/epidemiología , Antihelmínticos/uso terapéutico , Antihelmínticos/economía , Femenino , Masculino , Esquistosomiasis/diagnóstico , Esquistosomiasis/prevención & control , Esquistosomiasis/tratamiento farmacológico , Esquistosomiasis/epidemiología , Adulto , Adolescente , Niño , Quimioprevención/economía , Quimioprevención/métodos , Adulto Joven , Sensibilidad y Especificidad
15.
BMC Genomics ; 25(1): 192, 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38373909

RESUMEN

BACKGROUND: Control and elimination of schistosomiasis is an arduous task, with current strategies proving inadequate to break transmission. Exploration of genetic approaches to interrupt Schistosoma mansoni transmission, the causative agent for human intestinal schistosomiasis in sub-Saharan Africa and South America, has led to genomic research of the snail vector hosts of the genus Biomphalaria. Few complete genomic resources exist, with African Biomphalaria species being particularly underrepresented despite this being where the majority of S. mansoni infections occur. Here we generate and annotate the first genome assembly of Biomphalaria sudanica sensu lato, a species responsible for S. mansoni transmission in lake and marsh habitats of the African Rift Valley. Supported by whole-genome diversity data among five inbred lines, we describe orthologs of immune-relevant gene regions in the South American vector B. glabrata and present a bioinformatic pipeline to identify candidate novel pathogen recognition receptors (PRRs). RESULTS: De novo genome and transcriptome assembly of inbred B. sudanica originating from the shoreline of Lake Victoria (Kisumu, Kenya) resulted in a haploid genome size of ~ 944.2 Mb (6,728 fragments, N50 = 1.067 Mb), comprising 23,598 genes (BUSCO = 93.6% complete). The B. sudanica genome contains orthologues to all described immune genes/regions tied to protection against S. mansoni in B. glabrata, including the polymorphic transmembrane clusters (PTC1 and PTC2), RADres, and other loci. The B. sudanica PTC2 candidate immune genomic region contained many PRR-like genes across a much wider genomic region than has been shown in B. glabrata, as well as a large inversion between species. High levels of intra-species nucleotide diversity were seen in PTC2, as well as in regions linked to PTC1 and RADres orthologues. Immune related and putative PRR gene families were significantly over-represented in the sub-set of B. sudanica genes determined as hyperdiverse, including high extracellular diversity in transmembrane genes, which could be under pathogen-mediated balancing selection. However, no overall expansion in immunity related genes was seen in African compared to South American lineages. CONCLUSIONS: The B. sudanica genome and analyses presented here will facilitate future research in vector immune defense mechanisms against pathogens. This genomic/transcriptomic resource provides necessary data for the future development of molecular snail vector control/surveillance tools, facilitating schistosome transmission interruption mechanisms in Africa.


Asunto(s)
Biomphalaria , Esquistosomiasis mansoni , Animales , Humanos , Schistosoma mansoni/genética , Biomphalaria/genética , Transcriptoma , Genómica , Kenia
16.
Antimicrob Agents Chemother ; 68(3): e0143223, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38289079

RESUMEN

We previously performed a genome-wide association study (GWAS) to identify the genetic basis of praziquantel (PZQ) response in schistosomes, identifying two quantitative trait loci situated on chromosomes 2 and 3. We reanalyzed this GWAS using the latest (version 10) genome assembly showing that a single locus on chromosome 3, rather than two independent loci, determines drug response. These results reveal that PZQ response is monogenic and demonstrates the importance of high-quality genomic information.


Asunto(s)
Antihelmínticos , Esquistosomiasis mansoni , Animales , Praziquantel/farmacología , Praziquantel/uso terapéutico , Schistosoma mansoni/genética , Estudio de Asociación del Genoma Completo , Resistencia a Medicamentos , Esquistosomiasis mansoni/tratamiento farmacológico , Antihelmínticos/farmacología , Antihelmínticos/uso terapéutico
17.
Antimicrob Agents Chemother ; 68(7): e0011424, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38780260

RESUMEN

Schistosomiasis, a widespread parasitic disease caused by the blood fluke of the genus Schistosoma, affects over 230 million people, primarily in developing countries. Praziquantel, the sole drug currently approved for schistosomiasis treatment, demonstrates effectiveness against patent infections. A recent study highlighted the antiparasitic properties of amiodarone, an anti-arrhythmic drug, exhibiting higher efficacy than praziquantel against prepatent infections. This study assessed the efficacy of amiodarone and praziquantel, both individually and in combination, against Schistosoma mansoni through comprehensive in vitro and in vivo experiments. In vitro experiments demonstrated synergistic activity (fractional inhibitory concentration index ≤0.5) for combinations of amiodarone with praziquantel. In a murine model of schistosomiasis featuring prepatent infections, treatments involving amiodarone (200 or 400 mg/kg) followed by praziquantel (200 or 400 mg/kg) yielded a substantial reduction in worm burden (60%-70%). Given the low efficacy of praziquantel in prepatent infections, combinations of amiodarone with praziquantel may offer clinical utility in the treatment of schistosomiasis.


Asunto(s)
Amiodarona , Praziquantel , Schistosoma mansoni , Esquistosomiasis mansoni , Amiodarona/farmacología , Amiodarona/uso terapéutico , Animales , Praziquantel/farmacología , Praziquantel/uso terapéutico , Schistosoma mansoni/efectos de los fármacos , Ratones , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/parasitología , Femenino , Antihelmínticos/farmacología , Antihelmínticos/uso terapéutico , Sinergismo Farmacológico , Quimioterapia Combinada , Masculino , Modelos Animales de Enfermedad
18.
Anal Chem ; 96(16): 6311-6320, 2024 04 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
19.
Proc Biol Sci ; 291(2024): 20240449, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38864320

RESUMEN

The WHO aims to eliminate schistosomiasis as a public health problem by 2030. However, standard morbidity measures poorly correlate to infection intensities, hindering disease monitoring and evaluation. This is exacerbated by insufficient evidence on Schistosoma's impact on health-related quality of life (HRQoL). We conducted community-based cross-sectional surveys and parasitological examinations in moderate-to-high Schistosoma mansoni endemic communities in Uganda. We calculated parasitic infections and used EQ-5D instruments to estimate and compare HRQoL utilities in these populations. We further employed Tobit/linear regression models to predict HRQoL determinants. Two-thirds of the 560 participants were diagnosed with parasitic infection(s), 49% having S. mansoni. No significant negative association was observed between HRQoL and S. mansoni infection status/intensity. However, severity of pain urinating (ß = -0.106; s.e. = 0.043) and body swelling (ß = -0.326; s.e. = 0.005), increasing age (ß = -0.016; s.e. = 0.033), reduced socio-economic status (ß = 0.128; s.e. = 0.032), and being unemployed predicted lower HRQoL. Symptom severity and socio-economic status were better predictors of short-term HRQoL than current S. mansoni infection status/intensity. This is key to disentangling the link between infection(s) and short-term health outcomes, and highlights the complexity of correlating current infection(s) with long-term morbidity. Further evidence is needed on long-term schistosomiasis-associated HRQoL, health and economic outcomes to inform the case for upfront investments in schistosomiasis interventions.


Asunto(s)
Calidad de Vida , Schistosoma mansoni , Esquistosomiasis mansoni , Esquistosomiasis mansoni/epidemiología , Uganda/epidemiología , Humanos , Estudios Transversales , Femenino , Masculino , Animales , Schistosoma mansoni/fisiología , Adulto , Adolescente , Niño , Persona de Mediana Edad , Adulto Joven
20.
PLoS Pathog ; 18(3): e1010309, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35316298

RESUMEN

The eggs of the parasitic blood fluke, Schistosoma, are the main drivers of the chronic pathologies associated with schistosomiasis, a disease of poverty afflicting approximately 220 million people worldwide. Eggs laid by Schistosoma mansoni in the bloodstream of the host are encapsulated by vascular endothelial cells (VECs), the first step in the migration of the egg from the blood stream into the lumen of the gut and eventual exit from the body. The biomechanics associated with encapsulation and extravasation of the egg are poorly understood. We demonstrate that S. mansoni eggs induce VECs to form two types of membrane extensions during encapsulation; filopodia that probe eggshell surfaces and intercellular nanotubes that presumably facilitate VEC communication. Encapsulation efficiency, the number of filopodia and intercellular nanotubes, and the length of these structures depend on the egg's vitality and, to a lesser degree, its maturation state. During encapsulation, live eggs induce VEC contractility and membranous structures formation in a Rho/ROCK pathway-dependent manner. Using elastic hydrogels embedded with fluorescent microbeads as substrates to culture VECs, live eggs induce VECs to exert significantly greater contractile forces during encapsulation than dead eggs, which leads to 3D deformations on both the VEC monolayer and the flexible substrate underneath. These significant mechanical deformations cause the VEC monolayer tension to fluctuate with the eventual rupture of VEC junctions, thus facilitating egg transit out of the blood vessel. Overall, our data on the mechanical interplay between host VECs and the schistosome egg improve our understanding of how this parasite manipulates its immediate environment to maintain disease transmission.


Asunto(s)
Esquistosomiasis mansoni , Esquistosomiasis , Animales , Células Endoteliales , Humanos , Óvulo , Schistosoma mansoni , Esquistosomiasis mansoni/parasitología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA