Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 195
Filtrar
1.
Infect Dis Poverty ; 13(1): 65, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39256885

RESUMEN

BACKGROUND: Combating infectious diseases and halting biodiversity loss are intertwined challenges crucial to ensure global health. Biodiversity can constrain the spread of vector-borne pathogens circulation, necessitating a deeper understanding of ecological mechanisms underlying this pattern. Our study evaluates the relative importance of biodiversity and the abundance of Bulinus truncatus, a major intermediate host for the trematode Schistosoma haematobium on the circulation of this human pathogen at aquatic transmission sites. METHODS: We combined mathematical modelling and a molecular based empirical study to specifically assess the effect of co-infections between S. haematobium and other trematodes within their B. truncatus snail hosts; and B. truncatus abundance at transmission sites, on the production of S. haematobium infective cercariae stages released into the aquatic environment. RESULTS: Our modelling approach shows that more competitive trematode species exploiting B. truncatus as an intermediate host at the transmission site level leads to higher co-infection rates within snail hosts, subsequently reducing the production of S. haematobium cercariae. Conversely, an increase in B. truncatus abundance results in lower co-infection rates, and a higher proportion of S. haematobium cercariae released into the environment. Our empirical data from the field support these findings, indicating a significant negative effect of local trematode species richness (P-value = 0.029; AIC = 14.9) and co-infection rates (P-value = 0.02, AIC = 17.4) on the dominance of S. haematobium based on our GLMM models, while B. truncatus abundance positively influences S. haematobium dominance (P-value = 0.047, AIC = 20.1). CONCLUSIONS: Our study highlights the importance of biodiversity in influencing the transmission of S. haematobium through the effect of antagonistic interactions between trematodes within bulinid snail hosts. This effect intensifies when B. truncatus populations are low, promoting co-infections within snails. In line with the One Health concept, our results suggest that maintaining high level of freshwater biodiversity to sustain global trematode diversity at transmission sites can help reducing the circulation of Schistosoma species locally.


Asunto(s)
Interacciones Huésped-Parásitos , Schistosoma haematobium , Trematodos , Animales , Schistosoma haematobium/fisiología , Trematodos/fisiología , Humanos , Esquistosomiasis Urinaria/transmisión , Esquistosomiasis Urinaria/parasitología , Bulinus/parasitología , Caracoles/parasitología , Biodiversidad , Coinfección/parasitología , Modelos Teóricos , Cercarias/fisiología
2.
Infect Dis Poverty ; 13(1): 63, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39218903

RESUMEN

BACKGROUND: The control of schistosomiasis is particularly difficult in sub-Saharan Africa, which currently harbours 95% of this disease. The target population for preventive chemotherapy (PC) is expanded to all age group at risk of infection, thus increasing the demands of praziquantel (PZQ) tablets according to the new released guideline by World Health Organization. Due to the gap between available PZQ for PC and requirements, alternative approaches to assess endemicity of schistosomiasis in a community, are urgently needed for more quick and precise methods. We aimed to find out to which degree the infection status of snails can be used to guide chemotherapy against schistosomiasis. METHODS: We searched literature published from January 1991 to December 2022, that reported on the prevalence rates of Schistosoma mansoni, S. haematobium in the intermediate snails Biomphalaria spp. and Bulinus spp., respectively, and in humans. A random effect model for meta-analyses was used to calculate the pooled prevalence estimate (PPE), with heterogeneity assessed using I-squared statistic (I2), with correlation and regression analysis for the exploration of the relationship between human S. mansoni and S. haematobium infections and that in their specific intermediate hosts. RESULTS: Forty-seven publications comprising 59 field investigations were included. The pooled PPE of schistosomiasis, schistosomiasis mansoni and schistosomiasis haematobium in humans were 27.5% [95% confidence interval (CI): 24.0-31.1%], 25.6% (95% CI: 19.9-31.3%), and 28.8% (95% CI: 23.4-34.3%), respectively. The snails showed an overall infection rate of 8.6% (95% CI: 7.7-9.4%), with 12.1% (95% CI: 9.9-14.2%) in the Biomphalaria spp. snails and 6.9% (95% CI: 5.7-8.1%) in the Bulinus spp. snails. The correlation coefficient was 0.3 (95% CI: 0.01-0.5%, P < 0.05) indicating that the two variables, i.e. all intermediate host snails on the one hand and the human host on the other, were positively correlated. CONCLUSIONS: The prevalence rate of S. mansoni and S. haematobium is still high in endemic areas. Given the significant, positive correlation between the prevalence of schistosomes in humans and the intermediate snail hosts, more attention should be paid to programme integration of snail surveillance in future.


Asunto(s)
Biomphalaria , Schistosoma haematobium , Schistosoma mansoni , Esquistosomiasis Urinaria , Esquistosomiasis mansoni , Animales , Humanos , Prevalencia , Esquistosomiasis mansoni/epidemiología , Esquistosomiasis mansoni/prevención & control , Esquistosomiasis mansoni/parasitología , Esquistosomiasis Urinaria/epidemiología , Esquistosomiasis Urinaria/prevención & control , Esquistosomiasis Urinaria/parasitología , Schistosoma haematobium/fisiología , Schistosoma mansoni/fisiología , Biomphalaria/parasitología , Caracoles/parasitología , Bulinus/parasitología , África del Sur del Sahara/epidemiología
3.
Infect Dis Poverty ; 13(1): 62, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39198901

RESUMEN

BACKGROUND: Mali is known to be a schistosomiasis-endemic country with a limited supply of clean water. This has forced many communities to rely on open freshwater bodies for many human-water contact (HWC) activities. However, the relationship between contact with these water systems and the level of schistosome infection is currently receiving limited attention. This study assessed human-water interactions including cercarial emergence pattern and their influences on urinary schistosomiasis transmission in two communities in the Kayes district of Mali. METHODS: We carried out a parasitological study first in children in September 2021, then a cross-sectional study of quantitative observations of human-water contact activities in the population, and finally a study of snail infectivity at contact points in September 2022. The study took place in two communities, Fangouné Bamanan and Diakalèl in the Kayes region of western Mali. The chronobiological study focused on cercarial release from naturally infected snails. Released cercariae were molecularly genotyped by targeting the cox1 region, and the ITS and 18S ribosmal DNA gene (18S rDNA) regions of the DNA. Links between sociodemographic parameters, human water-contact points and hematuria were established using multivariate statistical analysis or the logistic regression model. RESULTS: The main factor predisposing the 97 participants to water contact was domestic activity (62.9%). Of the 378 snails collected at 14 sampling sites, 27 (7.1%) excreted schistosome cercariae, with 15.0% (19/126) at Fangouné Bamanan and 3.3% (8/252) at Diakalel. The release of Schistosoma cercariae shows three different patterns in Fangouné Bamanan: (i) an early release peak (6:00-8:00 AM), (ii) a mid-day release peak (10:00 AM-12:00 PM) and (iii) a double peak: (6:00-8:00 AM) and (6:00-8:00 PM) cercariae release; and two release patterns in Diakalel: early release (6:00-8:00 AM) and (ii) mid-day release (12:00-2:00 PM). All cercariae released during early diurnal (6:00-8:00 AM) or nocturnal emission patterns (6:00-8:00 PM) were hybrids parasite having an cox1 S. bovis or S. curassoni associated with an ITS and 18S rDNA of S. haematobium while the cercariae released during diurnal, or mid-day patterns (8:00 AM-6:00 PM) were pure S. haematobium. CONCLUSIONS: Our study showed that domestic activity is the main source of exposure in the Kayes region. Two and three cercariae emission patterns were observed at Diakalel and Fangouné Bamanan respectively. These results suggest that the parasite adapts to the human-water contact period in order to increase its infectivity.


Asunto(s)
Cercarias , Schistosoma haematobium , Esquistosomiasis Urinaria , Humanos , Malí/epidemiología , Animales , Esquistosomiasis Urinaria/transmisión , Esquistosomiasis Urinaria/epidemiología , Esquistosomiasis Urinaria/parasitología , Niño , Masculino , Cercarias/fisiología , Estudios Transversales , Femenino , Adolescente , Schistosoma haematobium/fisiología , Schistosoma haematobium/genética , Caracoles/parasitología , Preescolar , Adulto , Agua/parasitología
4.
PLoS Negl Trop Dis ; 18(6): e0011836, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38857289

RESUMEN

The geographical range of schistosomiasis is affected by the ecology of schistosome parasites and their obligate host snails, including their response to temperature. Previous models predicted schistosomiasis' thermal optimum at 21.7°C, which is not compatible with the temperature in sub-Saharan Africa (SSA) regions where schistosomiasis is hyperendemic. We performed an extensive literature search for empirical data on the effect of temperature on physiological and epidemiological parameters regulating the free-living stages of S. mansoni and S. haematobium and their obligate host snails, i.e., Biomphalaria spp. and Bulinus spp., respectively. We derived nonlinear thermal responses fitted on these data to parameterize a mechanistic, process-based model of schistosomiasis. We then re-cast the basic reproduction number and the prevalence of schistosome infection as functions of temperature. We found that the thermal optima for transmission of S. mansoni and S. haematobium range between 23.1-27.3°C and 23.6-27.9°C (95% CI) respectively. We also found that the thermal optimum shifts toward higher temperatures as the human water contact rate increases with temperature. Our findings align with an extensive dataset of schistosomiasis prevalence in SSA. The refined nonlinear thermal-response model developed here suggests a more suitable current climate and a greater risk of increased transmission with future warming for more than half of the schistosomiasis suitable regions with mean annual temperature below the thermal optimum.


Asunto(s)
Schistosoma haematobium , Schistosoma mansoni , Temperatura , Animales , Humanos , Schistosoma haematobium/fisiología , Schistosoma mansoni/fisiología , África del Sur del Sahara/epidemiología , Biomphalaria/parasitología , Esquistosomiasis/transmisión , Esquistosomiasis/epidemiología , Esquistosomiasis mansoni/transmisión , Esquistosomiasis mansoni/epidemiología , Bulinus/parasitología , Esquistosomiasis Urinaria/transmisión , Esquistosomiasis Urinaria/epidemiología , Prevalencia
5.
Acta Parasitol ; 69(1): 648-663, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38302641

RESUMEN

BACKGROUND: Trematode infections of the genus Schistosoma can induce physiological and behavioral changes in intermediate snail hosts. This is because the parasite consumes essential resources necessary for the host's survival, prompting hosts to adapt their behavior to maintain some level of fitness before parasite-induced mortality occurs. METHODS: In this study, the reproductive and biochemical parameters of Biomphalaria alexandrina and Bulinus truncatus were examined during the cercareal shedding stage of infection with Schistosoma mansoni and Schistosoma haematobium, respectively, compared with controls. RESULTS: The study revealed an infection rate of 34.7% for S. mansoni and 30.4% for S. haematobium. In B. alexandrina infected with S. mansoni, a survival rate of 65.2% was recorded, along with a mean prepatent period of 30.3 ± 1.41 days, a mean shedding duration of 14.2 ± 0.16 days, and a mean lifespan of 44.1 ± 0.24 days. Meanwhile, in B. truncatus infected with S. haematobium, a survival rate of 56.4% was observed, with a mean prepatent period of 44.3 ± 1.41 days, a mean shedding duration of 22.6 ± 2.7 days, and a mean lifespan of 66.9 ± 1.6 days. Feeding increased in both infected species of snails, while the net reproductive rate (Ro) of the infected snails decreased. Total antioxidant (TAO) and lipid peroxidation activity increased in the two infected snail species during shedding, while Glutathione-S-transferase levels decreased. Lipid peroxidase activity and nitrogen oxide levels significantly decreased in infected B. alexandrina and increased in infected Bulinus. Steroid hormone levels were elevated in infected Biomphalaria, whereas they were reduced in infected Bulinus. Comet assay parameters showed an increase in the two infected genera after infection compared to control snails, indicating genotoxic damage and histopathological damage was observed. CONCLUSIONS: These findings demonstrate that infection with larva species diverse biochemical, hormonal, genotoxic, and histopathological changes in the tissues responsible for fecundity and reproduction in B. alexandrina and B. truncates comparing with controls.


Asunto(s)
Biomphalaria , Bulinus , Interacciones Huésped-Parásitos , Schistosoma mansoni , Animales , Biomphalaria/parasitología , Schistosoma mansoni/fisiología , Bulinus/parasitología , Schistosoma haematobium/genética , Schistosoma haematobium/fisiología , Conducta Alimentaria , Cercarias/fisiología , Reproducción
6.
PLoS Negl Trop Dis ; 17(7): e0011487, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37459286

RESUMEN

BACKGROUND: Schistosomiasis and hookworm infection remain public health problems in large parts of sub-Saharan Africa. The epidemiology of schistosomiasis and hookworm was studied in seasonal transmission settings in the northern part of Côte d'Ivoire. METHODOLOGY: In August 2018, a cross-sectional study was conducted. Urine and stool samples were collected from 742 individuals aged 6-96 years in 16 localities from four departments in northern Côte d'Ivoire. Urine samples were examined by a filtration method for quantification of Schistosoma haematobium eggs. Stool samples were subjected to duplicate Kato-Katz thick smears and eggs of Schistosoma mansoni and soil-transmitted helminths (STHs) were counted. Additionally, a questionnaire was administered to determine demographic characteristics and to identify risk factors of schistosomiasis and STHs. Malacologic surveys were carried out at water points that are contacted by humans and animals. PRINCIPAL FINDINGS: The prevalence of schistosomiasis was very low. Only two cases of S. mansoni were found (0.3%, 95% confidence interval [CI]: 0.1-1.0%). The distribution of S. haematobium was focal, with cases found only in two departments; Ferkessédougou (5.4%, 95% CI: 2.5-9.9%) and Ouangolodougou (2.7%, 95% CI: 0.9-6.3%). Hookworm was the only STH species observed with a prevalence of 1.5% (95% CI: 0.8-2.8%). A higher risk of S. haematobium infection was observed in males compared to females, but the difference was not statistically significant (2.3% versus 1.3%, odds ratio [OR]: 1.5, 95% CI: 0.8-2.7). Participants aged 16-20 years showed the highest prevalence of S. haematobium. A total of 111 human- and animal-water contact points were identified at 47 water sources. Three potential intermediate host snails of schistosomes were collected; namely, Bulinus forskalii (n = 761), Bulinus truncatus (n = 205), and Biomphalaria pfeifferi (n = 1). Yet, only one specimen of Bu. truncatus was found to be shedding schistosome cercariae. CONCLUSIONS/SIGNIFICANCE: This study confirms very low transmission of schistosomiasis and hookworm in northern Côte d'Ivoire. The establishment and rigorous implementation of integrated surveillance-response systems could lead to the elimination of schistosomiasis and hookworm in this part of Côte d'Ivoire.


Asunto(s)
Infecciones por Uncinaria , Esquistosomiasis mansoni , Esquistosomiasis , Masculino , Femenino , Animales , Humanos , Estudios Transversales , Côte d'Ivoire/epidemiología , Prevalencia , Estaciones del Año , Esquistosomiasis/epidemiología , Schistosoma haematobium/fisiología , Bulinus , Infecciones por Uncinaria/epidemiología , Suelo/parasitología , Factores de Riesgo , Agua , Esquistosomiasis mansoni/epidemiología , Esquistosomiasis mansoni/parasitología , Heces/parasitología
7.
PLoS Negl Trop Dis ; 15(11): e0009083, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34788280

RESUMEN

BACKGROUND: Schistosomiasis remains a global-health problem with over 90% of its burden concentrated in Africa. Field studies reflect the complex ways in which socio-cultural and socio-economic variables, affect the distribution of Schistosoma infections across different populations. This review set out to systematically investigate and quantify the differences in Schistosoma infection burdens between males and females in Africa for two of the most prevalent Schistosoma species-Schistosoma mansoni and Schistosoma haematobium. METHODOLOGY: We searched (from inception to 11th March 2020) Embase, MEDLINE, PubMed, and Web of Science for relevant studies on schistosomiasis. We included studies that report S. mansoni and/or S. haematobium prevalence and/or intensity data distributed between males and females. We conducted meta-analyses on the male to female (M:F) prevalence of infection ratios. Subgroup analyses were performed according to study baseline prevalence, sample size and the lower and upper age limit of study participants. We also present a descriptive analysis of differential risk and intensity of infection across males and females. Evidence for differences in the prevalence of schistosomiasis infection between males and females is presented, stratified by Schistosoma species. RESULT: We identified 128 relevant studies, with over 200,000 participants across 23 countries. Of all the reported differences in the prevalence of infection between males and females, only 41% and 34% were statistically significant for S. mansoni and S. haematobium, respectively. Similar proportions of studies (27% and 34% for for S. haematobium and S. mansoni, respectively) of the reported differences in intensity of infection between males and females were statistically significant. The meta-analyses summarized a higher prevalence of infection in males; pooled random-effects weighted M:F prevalence of infection ratios were 1.20 (95% CI 1.11-1.29) for S. haematobium and 1.15 (95% CI 1.08-1.22) for S. mansoni. However, females are underrespresented in some of the studies. Additionally, there was significant heterogeneity across studies (Higgins I2 statistic (p-values < 0.001, I2values>95%)). Results of the subgroup analysis showed that the baseline prevalence influenced the M:F prevalence ratios for S. haematobium and S. mansoni, with higher M:F prevalence of infection ratios in settings with a lower baseline prevalence of infection. Across the studies, we identified four major risk factors associated with infection rates: occupational and recreational water contact, knowledge, socio-economic factors and demographic factors. The effect of these risk factors on the burden of infection in males and females varied across studies. CONCLUSIONS: We find evidence of differences in prevalence of infection between males and females which may reflect differences in gender norms and water contact activities, suggesting that policy changes at the regional level may help ameliorate gender-related disparities in schistosomiasis infection burden. Collecting, robustly analysing, and reporting, sex-disaggregated epidemiological data, is currently lacking, but would be highly informative for planning effective treatment programmes and establishing those most at risk of schistosomiasis infections.


Asunto(s)
Esquistosomiasis Urinaria/parasitología , Esquistosomiasis mansoni/parasitología , Factores Sexuales , África/epidemiología , Animales , Femenino , Humanos , Masculino , Factores de Riesgo , Schistosoma haematobium/genética , Schistosoma haematobium/fisiología , Schistosoma mansoni/genética , Schistosoma mansoni/fisiología , Esquistosomiasis Urinaria/epidemiología , Esquistosomiasis mansoni/epidemiología
8.
PLoS Negl Trop Dis ; 15(10): e0009806, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34610025

RESUMEN

BACKGROUND: Infectious disease risk is driven by three interrelated components: exposure, hazard, and vulnerability. For schistosomiasis, exposure occurs through contact with water, which is often tied to daily activities. Water contact, however, does not imply risk unless the environmental hazard of snails and parasites is also present in the water. By increasing reliance on hazardous activities and environments, socio-economic vulnerability can hinder reductions in exposure to a hazard. We aimed to quantify the contributions of exposure, hazard, and vulnerability to the presence and intensity of Schistosoma haematobium re-infection. METHODOLOGY/PRINCIPAL FINDINGS: In 13 villages along the Senegal River, we collected parasitological data from 821 school-aged children, survey data from 411 households where those children resided, and ecological data from all 24 village water access sites. We fit mixed-effects logistic and negative binomial regressions with indices of exposure, hazard, and vulnerability as explanatory variables of Schistosoma haematobium presence and intensity, respectively, controlling for demographic variables. Using multi-model inference to calculate the relative importance of each component of risk, we found that hazard (Æ©wi = 0.95) was the most important component of S. haematobium presence, followed by vulnerability (Æ©wi = 0.91). Exposure (Æ©wi = 1.00) was the most important component of S. haematobium intensity, followed by hazard (Æ©wi = 0.77). Model averaging quantified associations between each infection outcome and indices of exposure, hazard, and vulnerability, revealing a positive association between hazard and infection presence (OR = 1.49, 95% CI 1.12, 1.97), and a positive association between exposure and infection intensity (RR 2.59-3.86, depending on the category; all 95% CIs above 1). CONCLUSIONS/SIGNIFICANCE: Our findings underscore the linkages between social (exposure and vulnerability) and environmental (hazard) processes in the acquisition and accumulation of S. haematobium infection. This approach highlights the importance of implementing both social and environmental interventions to complement mass drug administration.


Asunto(s)
Reinfección/parasitología , Schistosoma haematobium/fisiología , Esquistosomiasis Urinaria/parasitología , Vulnerabilidad Social , Adolescente , Animales , Niño , Preescolar , Estudios Transversales , Femenino , Humanos , Estudios Longitudinales , Masculino , Reinfección/epidemiología , Reinfección/psicología , Población Rural/estadística & datos numéricos , Schistosoma haematobium/genética , Schistosoma haematobium/aislamiento & purificación , Esquistosomiasis Urinaria/epidemiología , Esquistosomiasis Urinaria/psicología , Senegal/epidemiología , Poblaciones Vulnerables/estadística & datos numéricos , Agua/parasitología
9.
PLoS Negl Trop Dis ; 15(9): e0009712, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34570777

RESUMEN

Schistosome parasites infect more than 200 million people annually, mostly in sub-Saharan Africa, where people may be co-infected with more than one species of the parasite. Infection risk for any single species is determined, in part, by the distribution of its obligate intermediate host snail. As the World Health Organization reprioritizes snail control to reduce the global burden of schistosomiasis, there is renewed importance in knowing when and where to target those efforts, which could vary by schistosome species. This study estimates factors associated with schistosomiasis risk in 16 villages located in the Senegal River Basin, a region hyperendemic for Schistosoma haematobium and S. mansoni. We first analyzed the spatial distributions of the two schistosomes' intermediate host snails (Bulinus spp. and Biomphalaria pfeifferi, respectively) at village water access sites. Then, we separately evaluated the relationships between human S. haematobium and S. mansoni infections and (i) the area of remotely-sensed snail habitat across spatial extents ranging from 1 to 120 m from shorelines, and (ii) water access site size and shape characteristics. We compared the influence of snail habitat across spatial extents because, while snail sampling is traditionally done near shorelines, we hypothesized that snails further from shore also contribute to infection risk. We found that, controlling for demographic variables, human risk for S. haematobium infection was positively correlated with snail habitat when snail habitat was measured over a much greater radius from shore (45 m to 120 m) than usual. S. haematobium risk was also associated with large, open water access sites. However, S. mansoni infection risk was associated with small, sheltered water access sites, and was not positively correlated with snail habitat at any spatial sampling radius. Our findings highlight the need to consider different ecological and environmental factors driving the transmission of each schistosome species in co-endemic landscapes.


Asunto(s)
Schistosoma haematobium/fisiología , Schistosoma mansoni/fisiología , Esquistosomiasis Urinaria/parasitología , Esquistosomiasis mansoni/parasitología , Adolescente , Adulto , Distribución Animal , Animales , Niño , Reservorios de Enfermedades/parasitología , Ecosistema , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ríos/parasitología , Población Rural/estadística & datos numéricos , Schistosoma haematobium/genética , Schistosoma haematobium/aislamiento & purificación , Schistosoma mansoni/genética , Schistosoma mansoni/aislamiento & purificación , Esquistosomiasis Urinaria/epidemiología , Esquistosomiasis Urinaria/transmisión , Esquistosomiasis mansoni/epidemiología , Esquistosomiasis mansoni/transmisión , Senegal/epidemiología , Caracoles/parasitología , Caracoles/fisiología , Adulto Joven
10.
PLoS Negl Trop Dis ; 15(5): e0009396, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33983965

RESUMEN

The stability of parasite populations is regulated by density-dependent processes occurring at different stages of their life cycle. In dioecious helminth infections, density-dependent fecundity is one such regulatory process that describes the reduction in egg production by female worms in high worm burden within-host environments. In human schistosomiasis, the operation of density-dependent fecundity is equivocal and investigation is hampered by the inaccessibility of adult worms that are located intravascularly. Current understanding is almost exclusively limited to data collected from two human autopsy studies conducted over 40 years ago, with subsequent analyses having reached conflicting conclusions. Whether egg production is regulated in a density-dependent manner is key to predicting the effectiveness of interventions targeting the elimination of schistosomiasis and to the interpretation of parasitological data collected during monitoring and evaluation activities. Here, we revisit density-dependent fecundity in the two most globally important human Schistosoma spp. using a statistical modelling approach that combines molecular inference on the number of parents/adult worms in individual human hosts with parasitological egg count data from mainland Tanzania and Zanzibar. We find a non-proportional relationship between S. haematobium egg counts and inferred numbers of female worms, providing the first clear evidence of density-dependent fecundity in this schistosome species. We do not find robust evidence for density-dependent fecundity in S. mansoni because of high sensitivity to some modelling assumptions and the lower statistical power of the available data. We discuss the strengths and limitations of our model-based analytical approach and its potential for improving our understanding of density dependence in schistosomiasis and other human helminthiases earmarked for elimination.


Asunto(s)
Fertilidad , Schistosoma haematobium/fisiología , Schistosoma mansoni/fisiología , Esquistosomiasis Urinaria/parasitología , Esquistosomiasis mansoni/parasitología , Animales , Femenino , Humanos , Masculino , Modelos Estadísticos , Recuento de Huevos de Parásitos , Schistosoma haematobium/genética , Schistosoma mansoni/genética , Tanzanía
11.
Front Immunol ; 12: 620657, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33737927

RESUMEN

HIV-1 infection disproportionately affects women in sub-Saharan Africa, where areas of high HIV-1 prevalence and Schistosoma haematobium endemicity largely overlap. Female genital schistosomiasis (FGS), an inflammatory disease caused by S. haematobium egg deposition in the genital tract, has been associated with prevalent HIV-1 infection. Elevated levels of the chemokines MIP-1α (CCL-3), MIP-1ß (CCL-4), IP-10 (CXCL-10), and IL-8 (CXCL-8) in cervicovaginal lavage (CVL) have been associated with HIV-1 acquisition. We hypothesize that levels of cervicovaginal cytokines may be raised in FGS and could provide a causal mechanism for the association between FGS and HIV-1. In the cross-sectional BILHIV study, specimens were collected from 603 female participants who were aged 18-31 years, sexually active, not pregnant and participated in the HPTN 071 (PopART) HIV-1 prevention trial in Zambia. Participants self-collected urine, and vaginal and cervical swabs, while CVLs were clinically obtained. Microscopy and Schistosoma circulating anodic antigen (CAA) were performed on urine. Genital samples were examined for parasite-specific DNA by PCR. Women with FGS (n=28), defined as a positive Schistosoma PCR from any genital sample were frequency age-matched with 159 FGS negative (defined as negative Schistosoma PCR, urine CAA, urine microscopy, and colposcopy imaging) women. Participants with probable FGS (n=25) (defined as the presence of either urine CAA or microscopy in combination with one of four clinical findings suggestive of FGS on colposcope-obtained photographs) were also included, for a total sample size of 212. The concentrations of 17 soluble cytokines and chemokines were quantified by a multiplex bead-based immunoassay. There was no difference in the concentrations of cytokines or chemokines between participants with and without FGS. An exploratory analysis of those women with a higher FGS burden, defined by ≥2 genital specimens with detectable Schistosoma DNA (n=15) showed, after adjusting for potential confounders, a higher Th2 (IL-4, IL-5, and IL-13) and pro-inflammatory (IL-15) expression pattern in comparison to FGS negative women, with differences unlikely to be due to chance (p=0.037 for IL-4 and p<0.001 for IL-5 after adjusting for multiple testing). FGS may alter the female genital tract immune environment, but larger studies in areas of varying endemicity are needed to evaluate the association with HIV-1 vulnerability.


Asunto(s)
Cuello del Útero/fisiología , Infecciones por VIH/inmunología , VIH-1/fisiología , Schistosoma haematobium/fisiología , Esquistosomiasis Urinaria/inmunología , Vagina/fisiología , Animales , Antígenos Helmínticos/orina , Estudios Transversales , Citocinas/metabolismo , Enfermedades Endémicas , Femenino , Glicoproteínas/orina , Infecciones por VIH/epidemiología , Proteínas del Helminto/orina , Humanos , Prevalencia , Esquistosomiasis Urinaria/epidemiología , Vagina/patología , Zambia/epidemiología
12.
Parasitol Res ; 120(5): 1755-1770, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33687566

RESUMEN

Cercarial emission of schistosomes is a determinant in the transmission to the definitive host and constitutes a good marker to identify which definitive host is responsible for transmission, mainly in introgressive hybridization situations. Our goal was to test the hypothesis that micro-mammals play a role in Schistosoma haematobium, S. bovis, and/or S. haematobium x S. bovis transmission. Small mammal sampling was conducted in seven semi-lacustrine villages of southern Benin. Among the 62 animals trapped, 50 individuals were investigated for Schistosoma adults and eggs: 37 Rattus rattus, 3 Rattus norvegicus, 9 Mastomys natalensis, and 1 Crocidura olivieri. Schistosoma adults were found in four R. rattus and two M. natalensis, with a local prevalence reaching 80% and 50%, respectively. Two cercarial chronotypes were found from Bulinus globosus experimentally infected with miracidia extracted from naturally infected M. natalensis: a late diurnal and nocturnal chronotype, and an early diurnal, late diurnal, and nocturnal chronotype. The cytochrome C oxidase subunit I mtDNA gene of the collected schistosomes (adults, miracidia, and cercariae) belonged to the S. bovis clade. Eleven internal transcribed spacer rDNA profiles were found; four belonged to S. bovis and seven to S. haematobium x S. bovis. These molecular results together with the observed multi-peak chronotypes add M. natalensis as a new host implicated in S. haematobium x S. bovis transmission. We discuss the origin of the new chronotypes which have become more complex with the appearance of several peaks in a 24-h day. We also discuss how the new populations of offspring may optimize intra-host ecological niche, host spectrum, and transmission time period.


Asunto(s)
Introgresión Genética , Murinae/parasitología , Schistosoma haematobium/fisiología , Schistosoma/fisiología , Esquistosomiasis/parasitología , Esquistosomiasis/transmisión , Animales , Benin , Bulinus/parasitología , Cercarias/genética , ADN Mitocondrial , ADN Ribosómico , Ecosistema , Femenino , Interacciones Huésped-Parásitos , Masculino , Tipificación Molecular , Prevalencia , Ratas , Schistosoma/genética , Schistosoma haematobium/genética , Esquistosomiasis Urinaria/parasitología , Esquistosomiasis Urinaria/transmisión , Musarañas/parasitología
13.
Lancet Planet Health ; 4(8): e330-e342, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32800151

RESUMEN

BACKGROUND: Schistosomiasis is a neglected tropical disease of global medical and veterinary importance. As efforts to eliminate schistosomiasis as a public health problem and interrupt transmission gather momentum, the potential zoonotic risk posed by livestock Schistosoma species via viable hybridisation in sub-Saharan Africa have been largely overlooked. We aimed to investigate the prevalence, distribution, and multi-host, multiparasite transmission cycle of Haematobium group schistosomiasis in Senegal, West Africa. METHODS: In this epidemiological study, we carried out systematic surveys in definitive hosts (humans, cattle, sheep, and goats) and snail intermediate hosts, in 2016-18, in two areas of Northern Senegal: Richard Toll and Lac de Guiers, where transmission is perennial; and Barkedji and Linguère, where transmission is seasonal. The occurrence and distribution of Schistosoma species and hybrids were assessed by molecular analyses of parasitological specimens obtained from the different hosts. Children in the study villages aged 5-17 years and enrolled in school were selected from school registers. Adults (aged 18-78 years) were self-selecting volunteers. Livestock from the study villages in both areas were also randomly sampled, as were post-mortem samples from local abattoirs. Additionally, five malacological surveys of snail intermediate hosts were carried out at each site in open water sources used by the communities and their animals. FINDINGS: In May to August, 2016, we surveyed 375 children and 20 adults from Richard Toll and Lac de Guiers, and 201 children and 107 adults from Barkedji and Linguère; in October, 2017, to January, 2018, we surveyed 386 children and 88 adults from Richard Toll and Lac de Guiers, and 323 children and 85 adults from Barkedji and Linguère. In Richard Toll and Lac de Guiers the prevalence of urogenital schistosomiasis in children was estimated to be 87% (95% CI 80-95) in 2016 and 88% (82-95) in 2017-18. An estimated 63% (in 2016) and 72% (in 2017-18) of infected children were shedding Schistosoma haematobium-Schistosoma bovis hybrids. In adults in Richard Toll and Lac de Guiers, the prevalence of urogenital schistosomiasis was estimated to be 79% (52-97) in 2016 and 41% (30-54) in 2017-18, with 88% of infected samples containing S haematobium-S bovis hybrids. In Barkedji and Linguère the prevalence of urogenital schistosomiasis in children was estimated to be 30% (23-38) in 2016 and 42% (35-49) in 2017-18, with the proportion of infected children found to be shedding S haematobium-S bovis hybrid miracidia much lower than in Richard Toll and Lac de Guiers (11% in 2016 and 9% in 2017-18). In adults in Barkedji and Linguère, the prevalence of urogenital schistosomiasis was estimated to be 26% (17-36) in 2016 and 47% (34-60) in 2017-18, with 10% of infected samples containing S haematobium-S bovis hybrids. The prevalence of S bovis in the sympatric cattle population of Richard Toll and the Lac de Guiers was 92% (80-99), with S bovis also found in sheep (estimated prevalence 14% [5-31]) and goats (15% [5-33]). In Barkedji and Linguère the main schistosome species in livestock was Schistosoma curassoni, with an estimated prevalence of 73% (48-93) in sheep, 84% (61-98) in goats and 8% (2-24) in cattle. S haematobium-S bovis hybrids were not found in livestock. In Richard Toll and Lac de Guiers 35% of infected Bulinus spp snail intermediate hosts were found to be shedding S haematobium-S bovis hybrids (68% shedding S haematobium; 17% shedding S bovis); however, no snails were found to be shedding S haematobium hybrids in Barkedji and Linguère (29% shedding S haematobium; 71% shedding S curassoni). INTERPRETATION: Our findings suggest that hybrids originate in humans via zoonotic spillover from livestock populations, where schistosomiasis is co-endemic. Introgressive hybridisation, evolving host ranges, and wider ecosystem contexts could affect the transmission dynamics of schistosomiasis and other pathogens, demonstrating the need to consider control measures within a One Health framework. FUNDING: Zoonoses and Emerging Livestock Systems programme (UK Biotechnology and Biological Sciences Research Council, UK Department for International Development, UK Economic and Social Research Council, UK Medical Research Council, UK Natural Environment Research Council, and UK Defence Science and Technology Laboratory).


Asunto(s)
Enfermedades de los Bovinos/epidemiología , Enfermedades de las Cabras/epidemiología , Schistosoma/fisiología , Esquistosomiasis/epidemiología , Esquistosomiasis/veterinaria , Enfermedades de las Ovejas/epidemiología , Caracoles/parasitología , Adolescente , Adulto , Anciano , Distribución Animal , Animales , Bovinos , Enfermedades de los Bovinos/parasitología , Enfermedades de los Bovinos/transmisión , Niño , Femenino , Enfermedades de las Cabras/parasitología , Enfermedades de las Cabras/transmisión , Cabras , Humanos , Masculino , Persona de Mediana Edad , Salud Única , Prevalencia , Schistosoma haematobium/fisiología , Esquistosomiasis/parasitología , Esquistosomiasis/transmisión , Senegal/epidemiología , Ovinos , Enfermedades de las Ovejas/parasitología , Enfermedades de las Ovejas/transmisión , Oveja Doméstica , Adulto Joven
14.
Am J Trop Med Hyg ; 103(1_Suppl): 80-91, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32400355

RESUMEN

Analyses of the population genetic structure of schistosomes under the "Schistosomiasis Consortium for Operational Research and Evaluation" (SCORE) contrasting treatment pressure scenarios in Tanzania, Niger, and Zanzibar were performed to provide supplementary critical information with which to evaluate the impact of these large-scale control activities and guide how activities could be adjusted. We predicted that population genetic analyses would reveal information on a range of important parameters including, but not exclusive to, recruitment and transmission of genotypes, occurrence of hybridization events, differences in reproductive mode, and degrees of inbreeding, and hence, the evolutionary potential, and responses of parasite populations under contrasting treatment pressures. Key findings revealed that naturally high levels of gene flow and mixing of the parasite populations between neighboring sites were likely to dilute any effects imposed by the SCORE treatment arms. Furthermore, significant inherent differences in parasite fecundity were observed, independent of current treatment arm, but potentially of major impact in terms of maintaining high levels of ongoing transmission in persistent "biological hotspot" sites. Within Niger, naturally occurring Schistosoma haematobium/Schistosoma bovis viable hybrids were found to be abundant, often occurring in significantly higher proportions than that of single-species S. haematobium infections. By examining parasite population genetic structures across hosts, treatment regimens, and the spatial landscape, our results to date illustrate key transmission processes over and above that which could be achieved through standard parasitological monitoring of prevalence and intensity alone, as well as adding to our understanding of Schistosoma spp. life history strategies in general.


Asunto(s)
Genética de Población , Schistosoma/genética , Esquistosomiasis/transmisión , África del Sur del Sahara/epidemiología , Animales , Antihelmínticos/uso terapéutico , Humanos , Hibridación Genética , Estadios del Ciclo de Vida , Administración Masiva de Medicamentos , Prevalencia , Schistosoma/efectos de los fármacos , Schistosoma/fisiología , Schistosoma haematobium/efectos de los fármacos , Schistosoma haematobium/genética , Schistosoma haematobium/fisiología , Esquistosomiasis/tratamiento farmacológico , Esquistosomiasis/epidemiología
15.
Parasit Vectors ; 13(1): 268, 2020 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-32448268

RESUMEN

BACKGROUND: Urogenital schistosomiasis, caused by infection with Schistosoma haematobium, is endemic in Niger but complicated by the presence of Schistosoma bovis, Schistosoma curassoni and S. haematobium group hybrids along with various Bulinus snail intermediate host species. Establishing the schistosomes and snails involved in transmission aids disease surveillance whilst providing insights into snail-schistosome interactions/compatibilities and biology. METHODS: Infected Bulinus spp. were collected from 16 villages north and south of the Niamey region, Niger, between 2011 and 2015. From each Bulinus spp., 20-52 cercariae shed were analysed using microsatellite markers and a subset identified using the mitochondrial (mt) cox1 and nuclear ITS1 + 2 and 18S DNA regions. Infected Bulinus spp. were identified using both morphological and molecular analysis (partial mt cox1 region). RESULTS: A total of 87 infected Bulinus from 24 sites were found, 29 were molecularly confirmed as B. truncatus, three as B. forskalii and four as B. globosus. The remaining samples were morphologically identified as B. truncatus (n = 49) and B. forskalii (n = 2). The microsatellite analysis of 1124 cercariae revealed 186 cercarial multilocus genotypes (MLGs). Identical cercarial genotypes were frequently (60%) identified from the same snail (clonal populations from a single miracidia); however, several (40%) of the snails had cercariae of different genotypes (2-10 MLG's) indicating multiple miracidial infections. Fifty-seven of the B. truncatus and all of the B. forskalii and B. globosus were shedding the Bovid schistosome S. bovis. The other B. truncatus were shedding the human schistosomes, S. haematobium (n = 6) and the S. haematobium group hybrids (n = 13). Two B. truncatus had co-infections with S. haematobium and S. haematobium group hybrids whilst no co-infections with S. bovis were observed. CONCLUSIONS: This study has advanced our understanding of human and bovid schistosomiasis transmission in the Niger River Valley region. Human Schistosoma species/forms (S. haematobium and S. haematobium hybrids) were found transmitted only in five villages whereas those causing veterinary schistosomiasis (S. bovis), were found in most villages. Bulinus truncatus was most abundant, transmitting all Schistosoma species, while the less abundant B. forskalii and B. globosus, only transmitted S. bovis. Our data suggest that species-specific biological traits may exist in relation to co-infections, snail-schistosome compatibility and intramolluscan schistosome development.


Asunto(s)
Bulinus/fisiología , Interacciones Huésped-Parásitos , Schistosoma haematobium/fisiología , Animales , Cercarias/genética , Cercarias/fisiología , Ciclooxigenasa 1/genética , Repeticiones de Microsatélite , Niger , Ríos , Schistosoma haematobium/genética , Esquistosomiasis Urinaria/parasitología , Esquistosomiasis Urinaria/transmisión , Especificidad de la Especie
16.
PLoS Negl Trop Dis ; 14(4): e0007951, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32240157

RESUMEN

Schistosomes are parasitic blood flukes that infect >200 million people around the world. Free-swimming larval stages penetrate the skin, invade a blood vessel, and migrate through the heart and lungs to the vasculature of the liver, where maturation and mating occurs. From here, the parasite couples migrate to their preferred egg laying sites. Here, we compare and contrast what is known about the migration patterns within the definitive host of the three major species of human schistosome: Schistosoma mansoni, S. japonicum, and S. haematobium. We conclude that intravascular schistosomes are inexorable colonizers whose migration and egg laying strategy is profligate; all three species (and their eggs) can be found throughout the mesenteric venules, the rectal venous plexus, and, to a greater or lesser extent, the urogenital venous plexuses. In addition, it is common for parasite eggs to be deposited in locations that lack easy access to the exterior, further demonstrating the relentless exploratory nature of these intravascular worms.


Asunto(s)
Vasos Sanguíneos/parasitología , Locomoción , Schistosoma haematobium/fisiología , Schistosoma japonicum/fisiología , Schistosoma mansoni/fisiología , Animales , Humanos , Estadios del Ciclo de Vida , Esquistosomiasis Urinaria/parasitología , Esquistosomiasis Japónica/parasitología , Esquistosomiasis mansoni/parasitología
18.
Parasitology ; 147(3): 287-294, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31727202

RESUMEN

Schistosomiasis is a neglected tropical disease, though it is highly prevalent in many parts of sub-Saharan Africa. While Schistosoma haematobium-bovis hybrids have been reported in West Africa, no data about Schistosoma hybrids in humans are available from Côte d'Ivoire. This study aimed to identify and quantify S. haematobium-bovis hybrids among schoolchildren in four localities of Côte d'Ivoire. Urine samples were collected and examined by filtration to detect Schistosoma eggs. Eggs were hatched and 503 miracidia were individually collected and stored on Whatman® FTA cards for molecular analysis. Individual miracidia were molecularly characterized by analysis of mitochondrial cox1 and nuclear internal transcribed spacer 2 (ITS 2) DNA regions. A mitochondrial cox1-based diagnostic polymerase chain reaction was performed on 459 miracidia, with 239 (52.1%) exhibiting the typical band for S. haematobium and 220 (47.9%) the S. bovis band. The cox1 and ITS 2 amplicons were Sanger sequenced from 40 randomly selected miracidia to confirm species and hybrids status. Among the 33 cox1 sequences analysed, we identified 15 S. haematobium sequences (45.5%) belonging to seven haplotypes and 18 S. bovis sequences (54.5%) belonging to 12 haplotypes. Of 40 ITS 2 sequences analysed, 31 (77.5%) were assigned to pure S. haematobium, four (10.0%) to pure S. bovis and five (12.5%) to S. haematobium-bovis hybrids. Our findings suggest that S. haematobium-bovis hybrids are common in Côte d'Ivoire. Hence, intense prospection of domestic and wild animals is warranted to determine whether zoonotic transmission occurs.


Asunto(s)
Hibridación Genética , Schistosoma/fisiología , Esquistosomiasis/epidemiología , Adolescente , Animales , Niño , Preescolar , Côte d'Ivoire/epidemiología , ADN de Helmintos/análisis , ADN Intergénico/análisis , Complejo IV de Transporte de Electrones/análisis , Proteínas del Helminto/análisis , Humanos , Proteínas Mitocondriales/análisis , Prevalencia , Schistosoma/genética , Schistosoma haematobium/genética , Schistosoma haematobium/fisiología , Esquistosomiasis/parasitología
19.
Parasit Vectors ; 12(1): 379, 2019 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-31358021

RESUMEN

BACKGROUND: Global changes promote the spread of infectious diseases worldwide. In this context, tropical urogenital schistosomiasis is now permanently established in Corsica since its first emergence in 2013. The local persistence of the tropical pathogens (schistosomes) responsible for urogenital schistosomiasis at such latitudes might be explained by (i) the presence of its intermediate host, the snail Bulinus truncatus, (ii) the recurrent local reseeding of schistosomes by their vertebrate hosts (either human or animal) every summer, and/or (iii) the maintenance and survival of schistosomes within their snail hosts over winter. METHODS: In this study we conducted an ecological experiment to assess the ability of temperate and tropical schistosome strains to survive in classical winter temperatures in Corsican rivers when infecting temperate (local) snail strains. We also quantified the ability of the schistosomes to complete their life-cycle post-overwintering when returned to classical summer water temperatures. RESULTS: Our results show that Mediterranean molluscs are locally adapted to winter conditions compared to tropical molluscs. Moreover, temperate and tropical schistosome strains equally survived the cold and produced viable offspring when returned to optimal temperatures. These results indicate that schistosomes can overwinter under temperate climates when infecting locally adapted snails and might partly explain the establishment and maintenance of schistosomes in Corsica from year to year. CONCLUSIONS: The observed broader thermal range of schistosomes compared to that of their snail hosts was unexpected and clearly indicates that the spread and establishment of schistosomiasis in temperate countries relies primarily on the presence of the locally adapted snail host lineages, currently known to be present in France, Italy, Portugal, Spain and Greece.


Asunto(s)
Adaptación Fisiológica , Frío , Schistosoma haematobium/fisiología , Esquistosomiasis Urinaria/epidemiología , Caracoles/fisiología , Caracoles/parasitología , Animales , Europa (Continente)/epidemiología , Francia/epidemiología , Interacciones Huésped-Parásitos , Esquistosomiasis Urinaria/parasitología , Esquistosomiasis Urinaria/transmisión , Estaciones del Año , Medicina Tropical
20.
PLoS Negl Trop Dis ; 13(5): e0007268, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31059495

RESUMEN

BACKGROUND: The Zanzibar Elimination of Schistosomiasis Transmission (ZEST) project aimed to eliminate urogenital schistosomiasis as a public health problem from Pemba and to interrupt Schistosoma haematobium transmission from Unguja in 5 years. METHODOLOGY: A repeated cross-sectional cluster-randomized trial was implemented from 2011/12 till 2017. On each island, 45 shehias were randomly assigned to receive one of three interventions: biannual mass drug administration (MDA) with praziquantel alone, or in combination with snail control or behavior change measures. In cross-sectional surveys, a single urine sample was collected from ~9,000 students aged 9- to 12-years and from ~4,500 adults aged 20- to 55-years annually, and from ~9,000 1st year students at baseline and the final survey. Each sample was examined for S. haematobium eggs by a single urine filtration. Prevalence and infection intensity were determined. Odds of infection were compared between the intervention arms. PRINCIPAL FINDINGS: Prevalence was reduced from 6.1% (95% confidence interval (CI): 4.5%-7.6%) to 1.7% (95% CI: 1.2%-2.2%) in 9- to 12-year old students, from 3.9% (95% CI: 2.8%-5.0%) to 1.5% (95% CI: 1.0%-2.0%) in adults, and from 8.8% (95% CI: 6.5%-11.2%) to 2.6% (95% CI: 1.7%-3.5%) in 1st year students from 2011/12 to 2017. In 2017, heavy infection intensities occurred in 0.4% of 9- to 12-year old students, 0.1% of adults, and 0.8% of 1st year students. Considering 1st year students in 2017, 13/45 schools in Pemba and 4/45 schools in Unguja had heavy infection intensities >1%. There was no significant difference in prevalence between the intervention arms in any study group and year. CONCLUSIONS/SIGNIFICANCE: Urogenital schistosomiasis was eliminated as public health problem from most sites in Pemba and Unguja. Prevalence was significantly reduced, but transmission was not interrupted. Continued interventions that are adaptive and tailored to the micro-epidemiology of S. haematobium in Zanzibar are needed to sustain and advance the gains made by ZEST.


Asunto(s)
Antihelmínticos/administración & dosificación , Praziquantel/administración & dosificación , Schistosoma haematobium/fisiología , Esquistosomiasis Urinaria/prevención & control , Adolescente , Adulto , Animales , Niño , Estudios Transversales , Erradicación de la Enfermedad , Femenino , Humanos , Islas del Oceano Índico/epidemiología , Islas/epidemiología , Masculino , Persona de Mediana Edad , Prevalencia , Schistosoma haematobium/efectos de los fármacos , Esquistosomiasis Urinaria/epidemiología , Esquistosomiasis Urinaria/parasitología , Caracoles/parasitología , Orina/parasitología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...