Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
Am J Hum Genet ; 107(3): 473-486, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32781046

RESUMEN

Africa contains more human genetic variation than any other continent, but the majority of the population-scale analyses of the African peoples have focused on just two of the four major linguistic groups, the Niger-Congo and Afro-Asiatic, leaving the Nilo-Saharan and Khoisan populations under-represented. In order to assess genetic variation and signatures of selection within a Nilo-Saharan population and between the Nilo-Saharan and Niger-Congo and Afro-Asiatic, we sequenced 50 genomes from the Nilo-Saharan Lugbara population of North-West Uganda and 250 genomes from 6 previously unsequenced Niger-Congo populations. We compared these data to data from a further 16 Eurasian and African populations including the Gumuz, another putative Nilo-Saharan population from Ethiopia. Of the 21 million variants identified in the Nilo-Saharan population, 3.57 million (17%) were not represented in dbSNP and included predicted non-synonymous mutations with possible phenotypic effects. We found greater genetic differentiation between the Nilo-Saharan Lugbara and Gumuz populations than between any two Afro-Asiatic or Niger-Congo populations. F3 tests showed that Gumuz contributed a genetic component to most Niger-Congo B populations whereas Lugabara did not. We scanned the genomes of the Lugbara for evidence of selective sweeps. We found selective sweeps at four loci (SLC24A5, SNX13, TYRP1, and UVRAG) associated with skin pigmentation, three of which already have been reported to be under selection. These selective sweeps point toward adaptations to the intense UV radiation of the Sahel.


Asunto(s)
Adaptación Fisiológica/genética , Variación Genética/genética , Selección Genética/genética , Pigmentación de la Piel/genética , Antiportadores/genética , Población Negra/genética , Manejo de Datos , Etiopía/epidemiología , Femenino , Genética de Población , Genoma Humano/genética , Haplotipos/genética , Humanos , Masculino , Glicoproteínas de Membrana/genética , Oxidorreductasas/genética , Polimorfismo de Nucleótido Simple/genética , Nexinas de Clasificación/genética , Proteínas Supresoras de Tumor/genética , Uganda/epidemiología
2.
PLoS Biol ; 17(1): e3000105, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30633739

RESUMEN

Human African trypanosomiasis (HAT), or African sleeping sickness, is a fatal disease found throughout sub-Saharan Africa. The disease is close to elimination in many areas, although it was similarly close to elimination once before and subsequently reemerged, despite seemingly low rates of transmission. Determining how these foci persisted and overcame an apparent transmission paradox is key to finally eliminating HAT. By assessing clinical, laboratory, and mathematical data, we propose that asymptomatic infections contribute to transmission through the presence of an overlooked reservoir of skin-dwelling parasites. Our assessment suggests that a combination of asymptomatic and parasitaemic cases is sufficient to maintain transmission at foci without animal reservoirs, and we argue that the current policy not to treat asymptomatic HAT should be reconsidered.


Asunto(s)
Tripanosomiasis Africana/etiología , Tripanosomiasis Africana/transmisión , África del Sur del Sahara/epidemiología , Animales , Infecciones Asintomáticas , Portador Sano/metabolismo , Humanos , Enfermedades Desatendidas/terapia , Tripanosomiasis Africana/tratamiento farmacológico
3.
Clin Infect Dis ; 73(1): 12-20, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32638003

RESUMEN

BACKGROUND: The diagnosis of gambiense human African trypanosomiasis (gHAT) typically involves 2 steps: a serological screen, followed by the detection of living trypanosome parasites in the blood or lymph node aspirate. Live parasites can, however, remain undetected in some seropositive individuals, who, we hypothesize, are infected with Trypanosoma brucei gambiense parasites in their extravascular dermis. METHODS: To test this hypothesis, we conducted a prospective observational cohort study in the gHAT focus of Forecariah, Republic of Guinea. Of the 5417 subjects serologically screened for gHAT, 66 were enrolled into our study and underwent a dermatological examination. At enrollment, 11 seronegative, 8 unconfirmed seropositive, and 18 confirmed seropositive individuals had blood samples and skin biopsies taken and examined for trypanosomes by molecular and immunohistological methods. RESULTS: In seropositive individuals, dermatological symptoms were significantly more frequent, relative to seronegative controls. T.b. gambiense parasites were present in the blood of all confirmed cases (n = 18) but not in unconfirmed seropositive individuals (n = 8). However, T. brucei parasites were detected in the extravascular dermis of all unconfirmed seropositive individuals and all confirmed cases. Skin biopsies of all treated cases and most seropositive untreated individuals progressively became negative for trypanosomes 6 and 20 months later. CONCLUSIONS: Our results highlight the skin as a potential reservoir for African trypanosomes, with implications for our understanding of this disease's epidemiology in the context of its planned elimination and underlining the skin as a novel target for gHAT diagnostics.


Asunto(s)
Tripanosomiasis Africana , Animales , Guinea , Humanos , Estudios Prospectivos , Trypanosoma brucei gambiense , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/epidemiología
4.
BMC Genomics ; 21(1): 289, 2020 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-32272904

RESUMEN

BACKGROUND: Copy number variation is an important class of genomic variation that has been reported in 75% of the human genome. However, it is underreported in African populations. Copy number variants (CNVs) could have important impacts on disease susceptibility and environmental adaptation. To describe CNVs and their possible impacts in Africans, we sequenced genomes of 232 individuals from three major African ethno-linguistic groups: (1) Niger Congo A from Guinea and Côte d'Ivoire, (2) Niger Congo B from Uganda and the Democratic Republic of Congo and (3) Nilo-Saharans from Uganda. We used GenomeSTRiP and cn.MOPS to identify copy number variant regions (CNVRs). RESULTS: We detected 7608 CNVRs, of which 2172 were only deletions, 2384 were only insertions and 3052 had both. We detected 224 previously un-described CNVRs. The majority of novel CNVRs were present at low frequency and were not shared between populations. We tested for evidence of selection associated with CNVs and also for population structure. Signatures of selection identified previously, using SNPs from the same populations, were overrepresented in CNVRs. When CNVs were tagged with SNP haplotypes to identify SNPs that could predict the presence of CNVs, we identified haplotypes tagging 3096 CNVRs, 372 CNVRs had SNPs with evidence of selection (iHS > 3) and 222 CNVRs had both. This was more than expected (p < 0.0001) and included loci where CNVs have previously been associated with HIV, Rhesus D and preeclampsia. When integrated with 1000 Genomes CNV data, we replicated their observation of population stratification by continent but no clustering by populations within Africa, despite inclusion of Nilo-Saharans and Niger-Congo populations within our dataset. CONCLUSIONS: Novel CNVRs in the current study increase representation of African diversity in the database of genomic variants. Over-representation of CNVRs in SNP signatures of selection and an excess of SNPs that both tag CNVs and are subject to selection show that CNVs may be the actual targets of selection at some loci. However, unlike SNPs, CNVs alone do not resolve African ethno-linguistic groups. Tag haplotypes for CNVs identified may be useful in predicting African CNVs in future studies where only SNP data is available.


Asunto(s)
Población Negra/genética , Variaciones en el Número de Copia de ADN , Genómica/métodos , África/etnología , Bases de Datos Genéticas , Predisposición Genética a la Enfermedad , Genética de Población , Genoma Humano , Haplotipos , Humanos
5.
Exp Parasitol ; 219: 108014, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33011238

RESUMEN

The objective set by WHO to reach elimination of human African trypanosomiasis (HAT) as a public health problem by 2020 is being achieved. The next target is the interruption of gambiense-HAT transmission in humans by 2030. To monitor progress towards this target, in areas where specialized local HAT control capacities will disappear, is a major challenge. Test specimens should be easily collectable and safely transportable such as dried blood spots (DBS). Monitoring tests performed in regional reference centres should be reliable, cheap and allow analysis of large numbers of specimens in a high-throughput format. The aim of this study was to assess the analytical sensitivity of Loopamp, M18S quantitative real-time PCR (M18S qPCR) and TgsGP qPCR as molecular diagnostic tests for the presence of Trypanosoma brucei gambiense in DBS. The sensitivity of the Loopamp test, with a detection limit of 100 trypanosomes/mL, was in the range of parasitaemias commonly observed in HAT patients, while detection limits for M18S and TgsGP qPCR were respectively 1000 and 10,000 trypanosomes/mL. None of the tests was entirely suitable for high-throughput use and further development and implementation of sensitive high-throughput molecular tools for monitoring HAT elimination are needed.


Asunto(s)
Técnicas de Diagnóstico Molecular/normas , Técnicas de Amplificación de Ácido Nucleico/normas , Reacción en Cadena en Tiempo Real de la Polimerasa/normas , Trypanosoma brucei gambiense/aislamiento & purificación , Tripanosomiasis Africana/prevención & control , Algoritmos , Animales , Recolección de Muestras de Sangre/métodos , Recolección de Muestras de Sangre/normas , ADN Protozoario/aislamiento & purificación , Ensayos Analíticos de Alto Rendimiento/métodos , Ensayos Analíticos de Alto Rendimiento/normas , Humanos , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Sensibilidad y Especificidad , Manejo de Especímenes/métodos , Manejo de Especímenes/normas , Trypanosoma brucei gambiense/genética , Tripanosomiasis Africana/sangre , Tripanosomiasis Africana/diagnóstico
6.
J Infect Dis ; 216(1): 22-28, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28586473

RESUMEN

Background: Visceral leishmaniasis (kala-azar, KA) is the most severe form of leishmaniasis, characterized by fever, weight loss, hepatosplenomegaly, and lymphadenopathy. During an outbreak of KA in Babar El Fugara (Sudan), 5.7% of cured patients displayed relapses, with familial clustering in half the cases. Methods: We performed whole-exome sequencing on 10 relapsing individuals and 11 controls from 5 nuclear families. Results: Rare homozygous and compound-heterozygous nonsense (c.1213C > T, rs139309795, p.Arg405*) and missense (c.701A > G, rs143439626, p.Lys234Arg) mutations of the alkylglycerol monooxygenase (AGMO) gene were associated with KA relapse in 3 families. Sequencing in additional family members confirmed the segregation of these mutations with relapse and revealed an autosomal dominant mode of transmission. These mutations were detected heterozygous in 2 subjects among 100 unrelated individuals with KA who never relapsed after cure, suggesting incomplete penetrance of AGMO deficiency. AGMO is expressed in hematopoietic cells, and is strongly expressed in the liver. AGMO modulates PAF production by mouse macrophages, suggesting that it may act through the PAF/PAF receptor pathway previously shown to have anti-Leishmania activity. Conclusions: This is the first demonstration that relapses after a first episode of KA are due to differences in human genetic susceptibility and not to modifications of parasite pathogenicity.


Asunto(s)
Exoma , Leishmaniasis Visceral/genética , Oxigenasas de Función Mixta/genética , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Estudios de Seguimiento , Predisposición Genética a la Enfermedad , Heterocigoto , Homocigoto , Humanos , Estudios Longitudinales , Masculino , Mutación , Recurrencia , Reproducibilidad de los Resultados , Sudán
7.
Clin Infect Dis ; 63(9): 1189-1197, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27470243

RESUMEN

BACKGROUND: Human African trypanosomiasis (HAT) caused by Trypanosoma brucei gambiense can be diagnosed in the early hemolymphatic stage (stage 1 [S1]) or meningoencephalitic stage (stage 2 [S2]). Importantly, individuals harbouring high and specific antibody responses to Tbg antigens but negative parasitology are also diagnosed in the field (seropositive [SERO]). Whereas some develop the disease in the months following their initial diagnosis (SERO/HAT), others remain parasitologically negative for long periods (SERO) and are apparently able to control infection. Human leucocyte antigen (HLA)-G, an immunosuppressive molecule, could play a critical role in this variability of progression between infection and disease. METHODS: Soluble HLA-G (sHLA-G) was measured in plasma for patients in the SERO (n = 65), SERO/HAT (n = 14), or HAT (n = 268) group and in cerebrospinal fluid for patients in S1 (n = 55), early S2 (n = 93), or late S2 (n = 110). Associations between these different statuses and the soluble level or genetic polymorphisms of HLA-G were explored. RESULTS: Plasma sHLA-G levels were significantly higher in HAT (P = 6 × 10-7) and SERO/HAT (P = .007) than SERO patients. No difference was observed between the SERO/HAT and HAT groups. Within the HAT group, specific haplotypes (HG010102 and HG0103) displayed increased frequencies in S1 (P = .013) and late S2 (P = .036), respectively. CONCLUSIONS: These results strongly suggest the involvement of HLA-G in HAT disease progression. Importantly, high plasma sHLA-G levels in SERO patients could be predictive of subsequent disease development and could represent a serological marker to help guide therapeutic decision making. Further studies are necessary to assess the predictive nature of HLA-G and to estimate both sensitivity and specificity.


Asunto(s)
Antígenos HLA-G/sangre , Tripanosomiasis Africana/sangre , Adulto , Biomarcadores/sangre , Progresión de la Enfermedad , Femenino , Haplotipos , Humanos , Masculino , Pronóstico , Trypanosoma brucei gambiense , Tripanosomiasis Africana/fisiopatología , Tripanosomiasis Africana/prevención & control
8.
PLoS Pathog ; 10(11): e1004469, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25375156

RESUMEN

In West Africa, Trypanosoma brucei gambiense, causing human African trypanosomiasis (HAT), is associated with a great diversity of infection outcomes. In addition to patients who can be diagnosed in the early hemolymphatic phase (stage 1) or meningoencephalitic phase (stage 2), a number of individuals can mount long-lasting specific serological responses while the results of microscopic investigations are negative (SERO TL+). Evidence is now increasing to indicate that these are asymptomatic subjects with low-grade parasitemia. The goal of our study was to investigate the type of immune response occurring in these "trypanotolerant" subjects. Cytokines levels were measured in healthy endemic controls (n = 40), stage 1 (n = 10), early stage 2 (n = 19), and late stage 2 patients (n = 23) and in a cohort of SERO TL+ individuals (n = 60) who were followed up for two years to assess the evolution of their parasitological and serological status. In contrast to HAT patients which T-cell responses appeared to be activated with increased levels of IL2, IL4, and IL10, SERO TL+ exhibited high levels of proinflammatory cytokines (IL6, IL8 and TNFα) and an almost absence of IL12p70. In SERO TL+, high levels of IL10 and low levels of TNFα were associated with an increased risk of developing HAT whereas high levels of IL8 predicted that serology would become negative. Further studies using high throughput technologies, hopefully will provide a more detailed view of the critical molecules or pathways underlying the trypanotolerant phenotype.


Asunto(s)
Inmunidad Innata , Interleucina-10/inmunología , Interleucina-8/inmunología , Trypanosoma brucei gambiense/inmunología , Tripanosomiasis Africana/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Interleucina-10/sangre , Interleucina-8/sangre , Masculino , Persona de Mediana Edad , Trypanosoma brucei gambiense/metabolismo , Tripanosomiasis Africana/sangre , Factor de Necrosis Tumoral alfa/sangre
9.
J Infect Dis ; 212(12): 1996-8, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26080371

RESUMEN

To assess the efficacy of treatment for human African trypanosomiasis, accurate tests that can discriminate relapse from cure are needed. We report the first data that the spliced leader (SL) RNA is a more specific marker for cure of human African trypanosomiasis than parasite DNA. In blood samples obtained from 61 patients in whom human African trypanosomiasis was cured, SL RNA detection had specificities of 98.4%-100%, while DNA detection had a specificity of only 77%. Data from our proof-of-concept study show that SL RNA detection has high potential as a test of cure.


Asunto(s)
ADN Protozoario/análisis , Monitoreo de Drogas/métodos , ARN Lider Empalmado/análisis , Trypanosoma brucei gambiense/aislamiento & purificación , Tripanosomiasis Africana/tratamiento farmacológico , ADN Protozoario/genética , Humanos , ARN Lider Empalmado/genética , Sensibilidad y Especificidad , Trypanosoma brucei gambiense/genética
10.
Trop Med Int Health ; 19(7): 828-31, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24750463

RESUMEN

OBJECTIVES: The immune trypanolysis test (TL) is an accurate sero-diagnostic tool increasingly implemented for sleeping sickness medical surveillance, but it is restricted to the reference laboratories. To facilitate storage and transport of the test specimen, we developed a protocol for the examination of blood spotted on filter paper (TL-fp) that can be stored and shipped at ambient temperature. We compared its performance with the classical TL on plasma (TL-pl) that needs to be kept frozen until use. METHODS: The study was conducted in active foci of the Republic of Guinea. In total, 438 specimens from treated and untreated sleeping sickness patients and serological suspects were tested with both methods. RESULT: TL-fp gave significantly less positive results than TL-pl, but all the confirmed sleeping sickness cases were positive with the TL-fp protocol. CONCLUSION: TL-fp appears to offer a good compromise between feasibility and sensitivity to detect currently infected subjects who play a role in the transmission of Trypanosoma brucei gambiense and is useful for contributing to the elimination of gambiense sleeping sickness.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Vigilancia de la Población/métodos , Trypanosoma brucei gambiense/inmunología , Tripanosomiasis Africana/epidemiología , Animales , Guinea/epidemiología , Humanos , Tamizaje Masivo/métodos , Enfermedades Desatendidas/epidemiología , Sensibilidad y Especificidad , Manejo de Especímenes/métodos , Tripanosomiasis Africana/sangre , Tripanosomiasis Africana/diagnóstico
11.
PLoS Negl Trop Dis ; 18(2): e0011985, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38377123

RESUMEN

Strategies to detect Human African Trypanosomiasis (HAT) cases rely on serological screening of populations exposed to trypanosomes. In Guinea, mass medical screening surveys performed with the Card Agglutination Test for Trypanosomiasis have been progressively replaced by door-to-door approaches using Rapid Diagnostic Tests (RDTs) since 2016. However, RDTs availability represents a major concern and medical teams must often adapt, even in the absence of prior RDT performance evaluation. For the last 5 years, the Guinean HAT National Control Program had to combine three different RDTs according to their availability and price: the SD Bioline HAT (not available anymore), the HAT Sero-K-SeT (most expensive), and recently the Abbott Bioline HAT 2.0 (limited field evaluation). Here, we assess the performance of these RDTs, alone or in different combinations, through the analysis of both prospective and retrospective data. A parallel assessment showed a higher positivity rate of Abbott Bioline HAT 2.0 (6.0%, n = 2,250) as compared to HAT Sero-K-SeT (1.9%), with a combined positive predictive value (PPV) of 20.0%. However, an evaluation of Abbott Bioline HAT 2.0 alone revealed a low PPV of 3.9% (n = 6,930) which was surpassed when using Abbott Bioline HAT 2.0 in first line and HAT Sero-K-SeT as a secondary test before confirmation, with a combined PPV reaching 44.4%. A retrospective evaluation of all 3 RDTs was then conducted on 189 plasma samples from the HAT-NCP biobank, confirming the higher sensitivity (94.0% [85.6-97.7%]) and lower specificity (83.6% [76.0-89.1%]) of Abbott Bioline HAT 2.0 as compared to SD Bioline HAT (Se 64.2% [52.2-74.6%]-Sp 98.4% [94.2-99.5%]) and HAT Sero-K-SeT (Se 88.1% [78.2-93.8%]-Sp 98.4% [94.2-99.5%]). A comparison of Abbott Bioline HAT 2.0 and malaria-RDT positivity rates on 479 subjects living in HAT-free malaria-endemic areas further revealed that a significantly higher proportion of subjects positive in Abbott Bioline HAT 2.0 were also positive in malaria-RDT, suggesting a possible cross-reaction of Abbott Bioline HAT 2.0 with malaria-related biological factors in about 10% of malaria cases. This would explain, at least in part, the limited specificity of Abbott Bioline HAT 2.0. Overall, Abbott Bioline HAT 2.0 seems suitable as first line RDT in combination with a second HAT RDT to prevent confirmatory lab overload and loss of suspects during referral for confirmation. A state-of-the-art prospective comparative study is further required for comparing all current and future HAT RDTs to propose an optimal combination of RDTs for door-to-door active screening.


Asunto(s)
Malaria , Tripanosomiasis Africana , Humanos , Animales , Tripanosomiasis Africana/diagnóstico , Papúa Nueva Guinea , Estudios Prospectivos , Estudios Retrospectivos
12.
Med Trop Sante Int ; 3(1)2023 03 31.
Artículo en Francés | MEDLINE | ID: mdl-37525637

RESUMEN

Human African Trypanosomiasis (HAT) is caused by Trypanosoma brucei which is transmitted by the tsetse fly insect vector (Glossina spp). It is one of the 20 Neglected Tropical Diseases (NTD) listed by the WHO. These diseases affect the poorest and most vulnerable communities, for which the WHO has established a dedicated 2021-2030 roadmap. At the time of Alphonse Laveran, HAT devastated the African continent. In the 1960s, the disease was nearly under control, but it strongly re-emerged in the 1990s. A coordinated effort of all stakeholders, with national control programs as the main actors, a strong contribution of research and important donations by the private sector, allowed to decrease the HAT burden significantly. Since 2018, less than 1000 cases are detected annually. We here review new diagnostics, treatments and vector control tools that have been implemented jointly and successfully in several endemic countries.The next key challenge will be to sustain the gains. Newly emerging research questions include long-term carriage of trypanosomes and adaptation of tools to low prevalence contexts. Challenges out of the research area comprise the continued need of funding, maintenance of dedicated human resources, and the key question of access. Sustainable elimination as "interruption of transmission", which is the 2030 NTD roadmap target, can be reached, if these challenges are solved. We stress the importance of continuing to combine the efforts in the fight against the disease, because sustainable elimination of HAT is the best long-term prevention strategy against re-emergence. As such, HAT elimination can serve as an example for other infectious diseases.


Asunto(s)
Trypanosoma brucei brucei , Tripanosomiasis Africana , Moscas Tse-Tse , Animales , Humanos , Tripanosomiasis Africana/epidemiología , Trypanosoma brucei gambiense , Insectos Vectores , Enfermedades Desatendidas/epidemiología
13.
Infect Dis Poverty ; 12(1): 22, 2023 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-36941656

RESUMEN

BACKGROUND: Passive diagnosis of human African trypanosomiasis (HAT) at the health facility level is a major component of HAT control in Guinea. We examined which clinical signs and symptoms are associated with HAT, and assessed the performance of selected clinical presentations, of rapid diagnostic tests (RDT), and of reference laboratory tests on dried blood spots (DBS) for diagnosing HAT in Guinea. METHOD: The study took place in 14 health facilities in Guinea, where 2345 clinical suspects were tested with RDTs (HAT Sero-K-Set, rHAT Sero-Strip, and SD Bioline HAT). Seropositives underwent parasitological examination (reference test) to confirm HAT and their DBS were tested in indirect enzyme-linked immunoassay (ELISA)/Trypanosoma brucei gambiense, trypanolysis, Loopamp Trypanosoma brucei Detection kit (LAMP) and m18S quantitative PCR (qPCR). Multivariable regression analysis assessed association of clinical presentation with HAT. Sensitivity, specificity, positive and negative predictive values of key clinical presentations, of the RDTs and of the DBS tests for HAT diagnosis were determined. RESULTS: The HAT prevalence, as confirmed parasitologically, was 2.0% (48/2345, 95% CI: 1.5-2.7%). Odds ratios (OR) for HAT were increased for participants with swollen lymph nodes (OR = 96.7, 95% CI: 20.7-452.0), important weight loss (OR = 20.4, 95% CI: 7.05-58.9), severe itching (OR = 45.9, 95% CI: 7.3-288.7) or motor disorders (OR = 4.5, 95% CI: 0.89-22.5). Presence of at least one of these clinical presentations was 75.6% (95% CI: 73.8-77.4%) specific and 97.9% (95% CI: 88.9-99.9%) sensitive for HAT. HAT Sero-K-Set, rHAT Sero-Strip, and SD Bioline HAT were respectively 97.5% (95% CI: 96.8-98.1%), 99.4% (95% CI: 99.0-99.7%) and 97.9% (95% CI: 97.2-98.4%) specific, and 100% (95% CI: 92.5-100.0%), 59.6% (95% CI: 44.3-73.3%) and 93.8% (95% CI: 82.8-98.7%) sensitive for HAT. The RDT's positive and negative predictive values ranged from 45.2-66.7% and 99.2-100% respectively. All DBS tests had specificities ≥ 92.9%. While LAMP and m18S qPCR sensitivities were below 50%, trypanolysis and ELISA/T.b. gambiense had sensitivities of 85.3% (95% CI: 68.9-95.0%) and 67.6% (95% CI: 49.5-82.6%). CONCLUSIONS: Presence of swollen lymph nodes, important weight loss, severe itching or motor disorders are simple but accurate clinical criteria for HAT referral in HAT endemic areas in Guinea. Diagnostic performances of HAT Sero-K-Set and SD Bioline HAT are sufficient for referring positives to microscopy. Trypanolysis on DBS may discriminate HAT patients from false RDT positives. Trial registration The trial was registered under NCT03356665 in clinicaltrials.gov (November 29, 2017, retrospectively registered https://clinicaltrials.gov/ct2/show/NCT03356665 ).


Asunto(s)
Tripanosomiasis Africana , Animales , Humanos , Pruebas Diagnósticas de Rutina , Guinea , Estudios Prospectivos , Sensibilidad y Especificidad
14.
Nat Commun ; 14(1): 7070, 2023 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-37923768

RESUMEN

In the skin, Trypanosoma brucei colonises the subcutaneous white adipose tissue, and is proposed to be competent for forward transmission. The interaction between parasites, adipose tissue, and the local immune system is likely to drive the adipose tissue wasting and weight loss observed in cattle and humans infected with T. brucei. However, mechanistically, events leading to subcutaneous white adipose tissue wasting are not fully understood. Here, using several complementary approaches, including mass cytometry by time of flight, bulk and single cell transcriptomics, and in vivo genetic models, we show that T. brucei infection drives local expansion of several IL-17A-producing cells in the murine WAT, including TH17 and Vγ6+ cells. We also show that global IL-17 deficiency, or deletion of the adipocyte IL-17 receptor protect from infection-induced WAT wasting and weight loss. Unexpectedly, we find that abrogation of adipocyte IL-17 signalling results in a significant accumulation of Dpp4+ Pi16+ interstitial preadipocytes and increased extravascular parasites in the WAT, highlighting a critical role for IL-17 signalling in controlling preadipocyte fate, subcutaneous WAT dynamics, and local parasite burden. Taken together, our study highlights the central role of adipocyte IL-17 signalling in controlling WAT responses to infection, suggesting that adipocytes are critical coordinators of tissue dynamics and immune responses to T. brucei infection.


Asunto(s)
Parásitos , Trypanosoma brucei brucei , Humanos , Ratones , Animales , Bovinos , Interleucina-17 , Tejido Adiposo , Grasa Subcutánea , Tejido Adiposo Blanco , Caquexia
15.
Infect Genet Evol ; 111: 105416, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36889485

RESUMEN

Eliminating schistosomiasis as a public health problem by 2030 requires a better understanding of the disease transmission, especially the asymmetric distribution of worm burden in individuals living and sharing the same environment. It is in this light that this study was designed to identify human genetic determinants associated with high burden of S. mansoni and also with the plasma concentrations of IgE and four cytokines in children from two schistosomiasis endemic areas of Cameroon. In school-aged children of schistosomiasis endemic areas of Makenene and Nom-Kandi of Cameroon, S. mansoni infections and their infection intensities were evaluated in urine and stool samples using respectively the Point-of-care Circulating Cathodic Antigen test (POC-CCA) and the Kato Katz (KK) test. Thereafter, blood samples were collected in children harbouring high burden of schistosome infections as well as in their parents and siblings. DNA extracts and plasma were obtained from blood. Polymorphisms at 14 loci of five genes were assessed using PCR-restriction fragment length polymorphism and amplification-refractory mutation system. The ELISA test enabled to determine the plasma concentrations of IgE, IL-13, IL-10, IL-4 and IFN-γ. The prevalence of S. mansoni infections was significantly higher (P < 0.0001 for POC-CCA; P = 0.001 for KK) in Makenene (48.6% for POC-CCA and 7.9% for KK) compared to Nom-Kandi (31% for POC-CCA and 4.3% for KK). The infection intensities were also higher (P < 0.0001 for POC-CCA; P = 0.001 for KK) in children from Makenene than those from Nom-Kandi. The allele C of SNP rs3024974 of STAT6 was associated with an increased risk of bearing high burden of S. mansoni both in the additive (p = 0.009) and recessive model (p = 0.01) while the allele C of SNP rs1800871 of IL10 was protective (p = 0.0009) against high burden of S. mansoni. The alleles A of SNP rs2069739 of IL13 and G of SNP rs2243283 of IL4 were associated with an increased risk of having low plasma concentrations of IL-13 (P = 0.04) and IL-10 (P = 0.04), respectively. This study showed that host genetic polymorphisms may influence the outcome (high or low worm burden) of S. mansoni infections and also the plasma concentrations of some cytokines.


Asunto(s)
Esquistosomiasis mansoni , Esquistosomiasis , Animales , Humanos , Niño , Schistosoma mansoni/genética , Interleucina-13/genética , Esquistosomiasis mansoni/epidemiología , Esquistosomiasis mansoni/genética , Interleucina-10/genética , Interleucina-4/genética , Citocinas/genética , Camerún/epidemiología , Antígenos Helmínticos/genética , Sensibilidad y Especificidad , Polimorfismo Genético , Prevalencia , Inmunoglobulina E , Heces
16.
Proc Natl Acad Sci U S A ; 106(1): 209-14, 2009 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19106297

RESUMEN

Human African trypanosomiasis, or sleeping sickness caused by Trypanosoma brucei gambiense, occurs in Western and Central Africa. T. brucei s.l. displays a huge diversity of adaptations and host specificities, and questions about its reproductive mode, dispersal abilities, and effective size remain under debate. We have investigated genetic variation at 8 microsatellite loci of T. b. gambiense strains isolated from human African trypanosomiasis patients in the Ivory Coast and Guinea, with the aim of knowing how genetic information was partitioned within and between individuals in both temporal and spatial scales. The results indicate that (i) migration of T. b. gambiense group 1 strains does not occur at the scale of West Africa, and that even at a finer scale (e.g., within Guinea) migration is restricted; (ii) effective population sizes of trypanosomes, as reflected by infected hosts, are probably higher than what the epidemiological surveys suggest; and (iii) T. b. gambiense group 1 is most likely a strictly clonally reproducing organism.


Asunto(s)
Variación Genética , Genética de Población , Trypanosoma brucei gambiense/genética , Tripanosomiasis Africana/parasitología , África Occidental , Animales , Côte d'Ivoire , Guinea , Humanos , Repeticiones de Microsatélite , Topografía Médica , Trypanosoma brucei gambiense/aislamiento & purificación , Tripanosomiasis Africana/epidemiología , Tripanosomiasis Africana/transmisión
17.
Vet Parasitol Reg Stud Reports ; 34: 100773, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36041808

RESUMEN

After intensive control efforts, human African trypanosomiasis (HAT) was declared eliminated in Côte d'Ivoire as a public health problem in December 2020 and the current objective is to achieve the interruption of the transmission (zero cases). Reaching this objective could be hindered by the existence of an animal reservoir of Trypanosoma (T.) brucei (b.) gambiense. In the framework of a study led in 2013 to assess the role of domestic animals in the epidemiology of HAT in the two last active foci from Côte d'Ivoire (Bonon and Sinfra), plasmas were sampled from four species of domestic animals for parasitological (microscopic examination by the buffy coat technique (BCT)), serological (immune trypanolysis (TL)) and molecular (specific PCR: TBR for T. brucei s.l., TCF for T. congolense forest type, TVW for T. vivax and PCR for T. b. gambiense) testing. In order to improve the understanding of the involvement/role of these animals in the transmission of T. b. gambiense, we have quantified in this study the IgG response to whole saliva extracts of Glossina palpalis gambiensis in order to perform an association analysis between anti-saliva responses and the positivity of diagnostic tests. Cattle and pigs had significantly higher rates of anti-tsetse saliva responses compared to goats and sheep (p < 0.01). In addition, the anti-tsetse saliva responses were strongly associated with the parasitology (BCT+), serology (TL+) and PCR (TBR+ and TCF+) results (p < 0.001). These associations indicate a high level of contacts between the positive/infected animals and tsetse flies. Our findings suggest that protecting cattle and pigs against tsetse bites could have a significant impact in reducing transmission of both animal and human trypanosome species, and advocates for a "One health" approach to better control African trypanosomosis in Côte d'Ivoire.


Asunto(s)
Enfermedades de los Bovinos , Enfermedades de las Ovejas , Enfermedades de los Porcinos , Trypanosoma , Tripanosomiasis Africana , Moscas Tse-Tse , Animales , Animales Domésticos , Formación de Anticuerpos , Bovinos , Enfermedades de los Bovinos/parasitología , Côte d'Ivoire/epidemiología , Humanos , Ovinos , Porcinos , Enfermedades de los Porcinos/parasitología , Tripanosomiasis Africana/epidemiología , Tripanosomiasis Africana/parasitología , Tripanosomiasis Africana/veterinaria , Moscas Tse-Tse/parasitología
18.
Parasite ; 29: 61, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36562442

RESUMEN

In the context of the human African trypanosomiasis elimination process, reliable and accurate diagnostic tools are crucial for exploring the role of a potential animal reservoir of Trypanosoma brucei gambiense. The immune trypanolysis test (TL) using the variant antigen types (VAT) LiTat 1.3 and LiTat 1.5, described as a specific serological method to detect people infected by T. b. gambiense, seems to be a promising tool. However, its specificity was recently questioned during field animal surveys. The present study evaluates the performance of TL during experimental T. b. brucei infection in pigs. Eight infected pigs and four uninfected pigs were followed up with blood and plasma collection. Blood was used for parasitological investigation. TL was performed on the plasma with the LiTat 1.3, LiTat 1.5 and LiTat 1.6 VATs. All control pigs remained negative to parasitological investigation and TL. Trypanosomes were detected in all the infected pigs and the first detection was between 10 and 14 days post infection (dpi). TL results showed that infected pigs developed antibodies against the three VATs. The first antibody detections by TL occurred between 14 and 21 dpi for antibodies directed against LiTat 1.6, 21 and 168 dpi for antibodies directed against LiTat 1.5 and 70, and 182 dpi for antibodies directed against LiTat 1.3. This study highlights for the first time that TL using LiTat 1.3 and LiTat 1.5 VATs is not specific to T. b. gambiense. Development of specific diagnostic tools for the detection of T. b. gambiense infections in animals, especially in pigs, is still needed.


Title: Évidence expérimentale que la trypanolyse basée sur les types d'antigène variable LiTat 1.3 et LiTat 1.5 n'est pas spécifique de Trypanosoma brucei gambiense. Abstract: Dans le contexte d'élimination de la trypanosomiase humaine Africaine, des outils de diagnostic fiables et précis sont essentiels afin d'explorer le rôle d'un potentiel réservoir animal de Trypanosoma brucei gambiense. La trypanolyse (TL) qui utilise les types d'antigène variable (TAV) LiTat 1.3 et LiTat 1.5, et qui est décrite comme une méthode sérologique spécifique pour détecter les personnes infectées par T. b. gambiense, semble être un outil prometteur. Cependant, sa spécificité a été récemment remise en question lors d'enquêtes sur les animaux. La présente étude évalue la performance de ce test lors d'une infection expérimentale à T. b. brucei chez le porc. Huit porcs infectés et quatre porcs témoins non infectés ont été suivis avec des prélèvements de sang et de plasma. Le sang a été utilisé pour l'examen parasitologique. La TL a été réalisée sur les échantillons de plasma avec les TAV LiTat 1.3, LiTat 1.5 et LiTat 1.6. Tous les porcs témoins ont été négatifs en parasitologie et à la TL. Les trypanosomes ont été détectés sur tous les porcs infectés avec les premières détections entre 10 et 14 jours post-infection (jpi). Les résultats de la TL ont montré que les porcs infectés ont développé des anticorps contre les trois TAV. Les premiers anticorps détectés par la TL étaient dirigés contre le LiTat 1.6 entre 14 et 21 jpi, puis le LiTat 1.5 entre 21 et 168 jpi et enfin le LiTat 1.3 entre 70 et 182 jpi. Cette étude démontre pour la première fois que la TL basée sur les TAV LiTat 1.3 et LiTat 1.5 n'est pas spécifique de T. b. gambiense. Il est donc toujours nécessaire et urgent de développer un outil de diagnostic spécifique pour la détection des infections à T. b. gambiense chez les animaux, notamment chez les porcs.


Asunto(s)
Trypanosoma brucei gambiense , Tripanosomiasis Africana , Animales , Humanos , Porcinos , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/veterinaria , Anticuerpos Antiprotozoarios
19.
PLoS Negl Trop Dis ; 15(2): e0009163, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33591980

RESUMEN

BACKGROUND: Activities to control human African trypanosomiasis (HAT) in Guinea were severely hampered by the Ebola epidemic that hit this country between 2014 and 2016. Active screening was completely interrupted and passive screening could only be maintained in a few health facilities. At the end of the epidemic, medical interventions were progressively intensified to mitigate the risk of HAT resurgence and progress towards disease elimination. METHODOLOGY/PRINCIPAL FINDINGS: A retrospective analysis was performed to evaluate the medical activities that were implemented in the three most endemic prefectures of Guinea (Boffa, Dubreka and Forecariah) between January 2016 and December 2018. Passive screening using rapid diagnostic tests (RDTs) was progressively resumed in one hundred and one health facilities, and active screening was intensified by visiting individual households and performing RDTs, and by conducting mass screening in villages by mobile teams using the Card Agglutination Test for Trypanosomiasis. A total of 1885, 4897 and 8023 clinical suspects were tested in passive, while 5743, 14442 and 21093 people were actively screened in 2016, 2017 and 2018, respectively. The number of HAT cases that were diagnosed first went up from 107 in 2016 to 140 in 2017, then subsequently decreased to only 73 in 2018. A progressive decrease in disease prevalence was observed in the populations that were tested in active and in passive between 2016 and 2018. CONCLUSIONS/SIGNIFICANCE: Intensified medical interventions in the post-Ebola context first resulted in an increase in the number of HAT cases, confirming the fear that the disease could resurge as a result of impaired control activities during the Ebola epidemic. On the other hand, the decrease in disease prevalence that was observed between 2016 and 2018 is encouraging, as it suggests that the current strategy combining enhanced diagnosis, treatment and vector control is appropriate to progress towards elimination of HAT in Guinea.


Asunto(s)
Tamizaje Masivo/estadística & datos numéricos , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/epidemiología , Pruebas Diagnósticas de Rutina/estadística & datos numéricos , Guinea/epidemiología , Fiebre Hemorrágica Ebola , Humanos , Prevalencia , Estudios Retrospectivos , Trypanosoma brucei gambiense/aislamiento & purificación
20.
AAS Open Res ; 4: 36, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35252746

RESUMEN

Background: Approximately 25% of the risk of Schistosoma mansoni is associated with host genetic variation. We will test 24 candidate genes, mainly in the T h2 and T h17 pathways, for association with S. mansoni infection intensity in four African countries, using family based and case-control approaches. Methods: Children aged 5-15 years will be recruited in S. mansoni endemic areas of Ivory Coast, Cameroon, Uganda and the Democratic Republic of Congo (DRC). We will use family based (study 1) and case-control (study 2) designs. Study 1 will take place in Ivory Coast, Cameroon, Uganda and the DRC. We aim to recruit 100 high worm burden families from each country except Uganda, where a previous study recruited at least 40 families. For phenotyping, cases will be defined as the 20% of children in each community with heaviest worm burdens as measured by the circulating cathodic antigen (CCA) assay. Study 2 will take place in Uganda. We will recruit 500 children in a highly endemic community. For phenotyping, cases will be defined as the 20% of children with heaviest worm burdens as measured by the CAA assay, while controls will be the 20% of infected children with the lightest worm burdens. Deoxyribonucleic acid (DNA) will be genotyped on the Illumina H3Africa SNP (single nucleotide polymorphisms) chip and genotypes will be converted to sets of haplotypes that span the gene region for analysis. We have selected 24 genes for genotyping that are mainly in the Th2 and Th17 pathways and that have variants that have been demonstrated to be or could be associated with Schistosoma infection intensity.   Analysis: In the family-based design, we will identify SNP haplotypes disproportionately transmitted to children with high worm burden. Case-control analysis will detect overrepresentation of haplotypes in extreme phenotypes with correction for relatedness by using whole genome principal components.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda