Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Parasite ; 31: 15, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38520091

RESUMO

Trypanosoma brucei gambiense (Tbg) group 2 is a subgroup of trypanosomes able to infect humans and is found in West and Central Africa. Unlike other agents causing sleeping sickness, such as Tbg group 1 and Trypanosoma brucei rhodesiense, Tbg2 lacks the typical molecular markers associated with resistance to human serum. Only 36 strains of Tbg2 have been documented, and therefore, very limited research has been conducted despite their zoonotic nature. Some of these strains are only available in their procyclic form, which hinders human serum resistance assays and mechanistic studies. Furthermore, the understanding of Tbg2's potential to infect tsetse flies and mammalian hosts is limited. In this study, 165 Glossina palpalis gambiensis flies were experimentally infected with procyclic Tbg2 parasites. It was found that 35 days post-infection, 43 flies out of the 80 still alive were found to be Tbg2 PCR-positive in the saliva. These flies were able to infect 3 out of the 4 mice used for blood-feeding. Dissection revealed that only six flies in fact carried mature infections in their midguts and salivary glands. Importantly, a single fly with a mature infection was sufficient to infect a mammalian host. This Tbg2 transmission success confirms that Tbg2 strains can establish in tsetse flies and infect mammalian hosts. This study describes an effective in vivo protocol for transforming Tbg2 from procyclic to bloodstream form, reproducing the complete Tbg2 cycle from G. p. gambiensis to mice. These findings provide valuable insights into Tbg2's host infectivity, and will facilitate further research on mechanisms of human serum resistance.


Title: Cycle de vie expérimental in vivo de Trypanosoma brucei gambiense groupe 2 : de la forme procyclique à la forme sanguicole. Abstract: Trypanosoma brucei gambiense (Tbg) groupe 2 est un sous-groupe de trypanosomes capables d'infecter l'Homme, présent en Afrique de l'Ouest et en Afrique centrale. Contrairement aux autres agents responsables de la maladie du sommeil, tels que Tbg groupe 1 et Trypanosoma brucei rhodesiense, Tbg2 ne présente pas les marqueurs moléculaires habituellement associés à la résistance au sérum humain. Seules trente-six souches de Tbg2 ont été répertoriées, limitant considérablement les recherches sur ce sous-groupe malgré sa nature zoonotique. Certaines de ces souches ne sont disponibles que sous leur forme procyclique, ce qui freine la réalisation des tests de résistance au sérum humain et les études mécanistiques. De plus, la compréhension du potentiel de Tbg2 à infecter les glossines et les hôtes mammifères est limitée. Dans cette étude, 165 glossines Glossina palpalis gambiensis ont été infectées expérimentalement par des parasites Tbg2 sous leur forme procyclique. Trente-cinq jours après l'infection, 43 des 80 glossines encore en vie se sont révélées positives à Tbg2 en PCR sur leur salive. Ces glossines ont réussi à infecter trois des quatre souris utilisées pour leur repas de sang. La dissection des glossines a révélé que seules six d'entre elles étaient réellement porteuses d'infections matures dans leur intestin et leurs glandes salivaires. Il est important de noter qu'une seule glossine porteuse d'une infection mature a suffi pour infecter un hôte mammifère. Ce succès de transmission de Tbg2 confirme que les souches de Tbg2 peuvent s'établir dans les glossines et infecter des hôtes mammifères. Cette étude décrit un protocole in vivo pour transformer la forme procyclique de Tbg2 en forme sanguicole, en reproduisant le cycle complet de Tbg2 de G. p. gambiensis à la souris. Ces résultats fournissent des informations précieuses sur le potentiel infectieux de Tbg2 et faciliteront la recherche sur les mécanismes de résistance au sérum humain des souches.


Assuntos
Trypanosoma brucei brucei , Trypanosoma , Tripanossomíase Africana , Moscas Tsé-Tsé , Animais , Humanos , Camundongos , Trypanosoma brucei gambiense , Tripanossomíase Africana/parasitologia , Moscas Tsé-Tsé/parasitologia , Estágios do Ciclo de Vida , Mamíferos
2.
PLoS Negl Trop Dis ; 18(2): e0011985, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38377123

RESUMO

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.


Assuntos
Malária , Tripanossomíase Africana , Humanos , Animais , Tripanossomíase Africana/diagnóstico , Papua Nova Guiné , Estudos Prospectivos , Estudos Retrospectivos
3.
Med Trop Sante Int ; 3(1)2023 03 31.
Artigo em Francês | MEDLINE | ID: mdl-37525637

RESUMO

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.


Assuntos
Trypanosoma brucei brucei , Tripanossomíase Africana , Moscas Tsé-Tsé , Animais , Humanos , Tripanossomíase Africana/epidemiologia , Trypanosoma brucei gambiense , Insetos Vetores , Doenças Negligenciadas/epidemiologia
4.
PLoS Negl Trop Dis ; 17(7): e0011528, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37498955

RESUMO

Although studies on African Trypanosomiases revealed a variety of trypanosome species in the blood of various animal taxa, animal reservoirs of Trypanosoma brucei gambiense and anatomical niches such as skin have been overlooked in most epidemiological settings. This study aims to update epidemiological data on trypanosome infections in animals from human African trypanosomiasis (HAT) foci of Cameroon. Blood and skin snips were collected from 291 domestic and wild animals. DNA was extracted from blood and skin snips and molecular approaches were used to identify different trypanosomes species. Immunohistochemical analyses were used to confirm trypanosome infections in skin snips. PCR revealed 137 animals (47.1%) with at least one trypanosome species in the blood and/or in the skin. Of these 137 animals, 90 (65.7%) and 32 (23.4%) had trypanosome infections respectively in the blood and skin. Fifteen (10.9%) animals had trypanosome infections in both blood and skin snip. Animals from the Campo HAT focus (55.0%) were significantly (X2 = 17.6; P< 0.0001) more infected than those (29.7%) from Bipindi. Trypanosomes of the subgenus Trypanozoon were present in 27.8% of animals while T. vivax, T. congolense forest type and savannah type were detected in 16.5%, 10.3% and 1.4% of animals respectively. Trypanosoma b. gambiense infections were detected in the blood of 7.6% (22/291) of animals. No T. b. gambiense infection was detected in skin. This study highlights the presence of several trypanosome species in the blood and skin of various wild and domestic animals. Skin appeared as an anatomical reservoir for trypanosomes in animals. Despite methodological limitations, pigs, sheep, goats and wild animals were confirmed as potential reservoirs of T. b. gambiense. These animal reservoirs must be considered for the designing of control strategies that will lead to sustainable elimination of HAT.


Assuntos
Trypanosoma , Tripanossomíase Africana , Moscas Tsé-Tsé , Humanos , Animais , Suínos , Ovinos , Tripanossomíase Africana/epidemiologia , Tripanossomíase Africana/veterinária , Camarões/epidemiologia , Prevalência , DNA de Protozoário/genética , DNA de Protozoário/química , Trypanosoma/genética , Trypanosoma brucei gambiense/genética , Animais Selvagens , Cabras , Moscas Tsé-Tsé/genética
5.
BMC Vet Res ; 18(1): 64, 2022 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-35120506

RESUMO

BACKGROUND: Brucellosis, Q fever and Rift Valley fever are considered as Neglected Zoonotic Diseases (NZDs) leading to socioeconomic losses in livestock globally, and particularly in developing countries of Africa where they are under-reported. In this study, we evaluated the seroprevalence of these 3 zoonotic diseases in domestic ruminants in Guinea from 2017 to 2019. A total of 1357 sera, sampled from 463 cattle, 408 goats and 486 sheep, were collected in 17 Guinean prefectures and analyzed by enzyme-linked immunosorbent assay (ELISA). RESULTS: Cattle was the species with highest seroprevalence (5 to 20-fold higher than in small ruminants) for the three diseases. The seroprevalence of brucellosis, mostly focused in Western Guinea, was 11.0% (51 of 463) in cattle, 0.4% (2 in 486) in sheep while no specific antibodies were found in goats. Q fever, widespread across the country, was the most frequently detected zoonosis with a mean seroprevalence of 20.5% (95 in 463), 4.4% (18 in 408) and 2.3% (11 in 486) in cattle, goats and sheep, respectively. The mean seroprevalence of RVF was 16.4% (76 in 463) in cattle, 1.0% (4 in 408) in goats and 1.0% (5 in 486) in sheep. Among the samples 19.3% were seropositive for at least one of the three NZDs, 2.5% showed specific antibodies against at least two pathogens and 4 cattle (0.8%) were seropositive for all three pathogens. In cattle, adults over 3-years old and females presented a higher antibody seroprevalence for the three diseases, in congruence with putative exposure risk. CONCLUSIONS: This study confirms the circulation of these three zoonotic pathogens in Guinea and highlights the need for implementing a syndromic surveillance of ruminant abortions by the Guinean veterinary authorities as well as for the screening of the human population at risk (veterinarians, breeders, slaughterers) in a One Health perspective.


Assuntos
Brucelose , Doenças das Cabras , Febre Q , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Doenças dos Ovinos , Aborto Animal , Animais , Brucelose/epidemiologia , Brucelose/veterinária , Bovinos , Ensaio de Imunoadsorção Enzimática/veterinária , Feminino , Doenças das Cabras/epidemiologia , Cabras , Guiné , Gravidez , Febre Q/epidemiologia , Febre Q/veterinária , Ruminantes , Estudos Soroepidemiológicos , Ovinos , Doenças dos Ovinos/epidemiologia
6.
Emerg Infect Dis ; 28(2): 457-460, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34906290

RESUMO

This overview of severe acute respiratory syndrome coronavirus 2 circulation over 1.5 years in Guinea demonstrates that virus clades and variants of interest and concern were progressively introduced, mostly by travellers through Conakry, before spreading through the country. Sequencing is key to following virus evolution and establishing efficient control strategies.


Assuntos
COVID-19 , SARS-CoV-2 , Guiné/epidemiologia , Humanos
7.
PLoS Negl Trop Dis ; 15(12): e0010036, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34937054

RESUMO

BACKGROUND: The existence of an animal reservoir of Trypanosoma brucei gambiense (T. b. gambiense), the agent of human African trypanosomiasis (HAT), may compromise the interruption of transmission targeted by World Health Organization. The aim of this study was to investigate the presence of trypanosomes in pigs and people in the Vavoua HAT historical focus where cases were still diagnosed in the early 2010's. METHODS: For the human survey, we used the CATT, mini-anion exchange centrifugation technique and immune trypanolysis tests. For the animal survey, the buffy coat technique was also used as well as the PCR using Trypanosoma species specific, including the T. b. gambiense TgsGP detection using single round and nested PCRs, performed from animal blood samples and from strains isolated from subjects positive for parasitological investigations. RESULTS: No HAT cases were detected among 345 people tested. A total of 167 pigs were investigated. Free-ranging pigs appeared significantly more infected than pigs in pen. Over 70% of free-ranging pigs were positive for CATT and parasitological investigations and 27-43% were positive to trypanolysis depending on the antigen used. T. brucei was the most prevalent species (57%) followed by T. congolense (24%). Blood sample extracted DNA of T. brucei positive subjects were negative to single round TgsGP PCR. However, 1/22 and 6/22 isolated strains were positive with single round and nested TgsGP PCRs, respectively. DISCUSSION: Free-ranging pigs were identified as a multi-reservoir of T. brucei and/or T. congolense with mixed infections of different strains. This trypanosome diversity hinders the easy and direct detection of T. b. gambiense. We highlight the lack of tools to prove or exclude with certainty the presence of T. b. gambiense. This study once more highlights the need of technical improvements to explore the role of animals in the epidemiology of HAT.


Assuntos
Reservatórios de Doenças/parasitologia , Doenças dos Suínos/parasitologia , Trypanosoma brucei gambiense/isolamento & purificação , Trypanosoma congolense/isolamento & purificação , Tripanossomíase Africana/parasitologia , Tripanossomíase Africana/veterinária , Animais , Animais Domésticos/parasitologia , Côte d'Ivoire/epidemiologia , Humanos , Reação em Cadeia da Polimerase , Suínos , Doenças dos Suínos/epidemiologia , Trypanosoma brucei gambiense/genética , Trypanosoma brucei gambiense/fisiologia , Trypanosoma congolense/genética , Trypanosoma congolense/fisiologia , Tripanossomíase Africana/epidemiologia
8.
PLoS Negl Trop Dis ; 15(3): e0009246, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33661894

RESUMO

One health (OH) approaches have increasingly been used in the last decade in the fight against zoonotic neglected tropical diseases (NTDs). However, descriptions of such collaborations between the human, animal and environmental health sectors are still limited for French-speaking tropical countries. The objective of the current survey was to explore the diversity of OH experiences applied to research, surveillance and control of NTDs by scientists from French-speaking countries, and discuss their constraints and benefits. Six zoonotic NTDs were targeted: echinococcoses, trypanosomiases, leishmaniases, rabies, Taenia solium cysticercosis and leptospiroses. Invitations to fill in an online questionnaire were sent to members of francophone networks on NTDs and other tropical diseases. Results from the questionnaire were discussed during an international workshop in October 2019. The vast majority (98%) of the 171 respondents considered OH approaches relevant although only 64% had implemented them. Among respondents with OH experience, 58% had encountered difficulties mainly related to a lack of knowledge, interest and support for OH approaches by funding agencies, policy-makers, communities and researchers. Silos between disciplines and health sectors were still strong at both scientific and operational levels. Benefits were reported by 94% of respondents with OH experience, including increased intellectual stimulation, stronger collaborations, higher impact and cost-efficiency of interventions. Recommendations for OH uptake included advocacy, capacity-building, dedicated funding, and higher communities' involvement. Improved research coordination by NTD networks, production of combined human-animal health NTD impact indicators, and transversal research projects on diagnostic and reservoirs were also considered essential.


Assuntos
Doenças Negligenciadas/prevenção & controle , Medicina Tropical , Zoonoses/prevenção & controle , Animais , Pesquisa Biomédica , Humanos , Inquéritos e Questionários
9.
Clin Infect Dis ; 73(1): 12-20, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32638003

RESUMO

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.


Assuntos
Tripanossomíase Africana , Animais , Guiné , Humanos , Estudos Prospectivos , Trypanosoma brucei gambiense , Tripanossomíase Africana/diagnóstico , Tripanossomíase Africana/epidemiologia
10.
PLoS Negl Trop Dis ; 14(11): e0008738, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33180776
11.
Pathogens ; 9(4)2020 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-32295095

RESUMO

For clinical epidemiology specialists, connecting the genetic diversity of Echinococcus multilocularis to sources of infection or particular sites has become somewhat of a holy grail. It is very difficult to trace the infection history of alveolar echinococcosis (AE) patients as there may be an incubation period of five to 15 years before reliable diagnosis. Moreover, the variability of parasitic manifestations in human patients raises the possibility of genetically different isolates of E. multilocularis having different levels of pathogenicity. Thus, the exposure of human patients to different strains or genotypes circulating in geographically different environments may lead to different disease outcomes. Molecular tools, such as the microsatellite marker EmsB, were required to investigate these aspects. This genetic marker was previously tested on a collection of 1211 European field samples predominantly of animal origin, referenced on a publicly available database. In this study, we investigated a panel of 66 metacestode samples (between 1981 and 2019) recovered surgically from 63 patients diagnosed with alveolar echinococcosis originating from four European countries (France, Switzerland, Germany, Belgium). In this study, we identified nine EmsB profiles, five of which were found in patients located in the same areas of France and Switzerland. One profile was detected on both sides of the French-Swiss border, whereas most patients from non-endemic regions clustered together in another profile. EmsB profiles appeared to remain stable over time because similar profiles were detected in patients who underwent surgery recently and patients who underwent surgery some time ago. This study sheds light on possible pathways of contamination in humans, including proximity contamination in some cases, and the dominant contamination profiles in Europe, particularly for extrahepatic lesions.

12.
Nat Commun ; 11(1): 1326, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32165615

RESUMO

Persistent pathogens have evolved to avoid elimination by the mammalian immune system including mechanisms to evade complement. Infections with African trypanosomes can persist for years and cause human and animal disease throughout sub-Saharan Africa. It is not known how trypanosomes limit the action of the alternative complement pathway. Here we identify an African trypanosome receptor for mammalian factor H, a negative regulator of the alternative pathway. Structural studies show how the receptor binds ligand, leaving inhibitory domains of factor H free to inactivate complement C3b deposited on the trypanosome surface. Receptor expression is highest in developmental stages transmitted to the tsetse fly vector and those exposed to blood meals in the tsetse gut. Receptor gene deletion reduced tsetse infection, identifying this receptor as a virulence factor for transmission. This demonstrates how a pathogen evolved a molecular mechanism to increase transmission to an insect vector by exploitation of a mammalian complement regulator.


Assuntos
Fator H do Complemento/metabolismo , Trypanosoma/fisiologia , Moscas Tsé-Tsé/parasitologia , Animais , Anticorpos Monoclonais/metabolismo , Células CHO , Bovinos , Membrana Celular/metabolismo , Complemento C3b/metabolismo , Fator H do Complemento/química , Cricetinae , Cricetulus , Camundongos Endogâmicos BALB C , Parasitemia/sangue , Ligação Proteica , Domínios Proteicos , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Receptores de Superfície Celular/metabolismo , Regulação para Cima
13.
Int J Parasitol Drugs Drug Resist ; 8(2): 203-212, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29649664

RESUMO

The parasitic protozoan Trypanosoma brucei is the causative agent of human African trypanosomiasis (sleeping sickness) and nagana. Current drug therapies have limited efficacy, high toxicity and/or are continually hampered by the appearance of resistance. Antimicrobial peptides have recently attracted attention as potential parasiticidal compounds. Here, we explore circular bacteriocin AS-48's ability to kill clinically relevant bloodstream forms of T. brucei gambiense, T. brucei rhodesiense and T. brucei brucei. AS-48 exhibited excellent anti-trypanosomal activity in vitro (EC50 = 1-3 nM) against the three T. brucei subspecies, but it was innocuous to human cells at 104-fold higher concentrations. In contrast to its antibacterial action, AS-48 does not kill the parasite through plasma membrane permeabilization but by targeting intracellular compartments. This was evidenced by the fact that vital dye internalization-prohibiting concentrations of AS-48 could kill the parasite at 37 °C but not at 4 °C. Furthermore, AS-48 interacted with the surface of the parasite, at least in part via VSG, its uptake was temperature-dependent and clathrin-depleted cells were less permissive to the action of AS-48. The bacteriocin also caused the appearance of myelin-like structures and double-membrane autophagic vacuoles. These changes in the parasite's ultrastructure were confirmed by fluorescence microscopy as AS-48 induced the production of EGFP-ATG8.2-labeled autophagosomes. Collectively, these results indicate AS-48 kills the parasite through a mechanism involving clathrin-mediated endocytosis of VSG-bound AS-48 and the induction of autophagic-like cell death. As AS-48 has greater in vitro activity than the drugs currently used to treat T. brucei infection and does not present any signs of toxicity in mammalian cells, it could be an attractive lead compound for the treatment of sleeping sickness and nagana.


Assuntos
Autofagia/efeitos dos fármacos , Bacteriocinas/farmacologia , Morte Celular/efeitos dos fármacos , Tripanossomicidas/farmacologia , Trypanosoma brucei brucei/efeitos dos fármacos , Animais , Endocitose/efeitos dos fármacos , Humanos , Microscopia de Fluorescência , Temperatura , Trypanosoma brucei brucei/ultraestrutura , Trypanosoma brucei gambiense/efeitos dos fármacos , Trypanosoma brucei rhodesiense/efeitos dos fármacos , Tripanossomíase Africana/sangue , Tripanossomíase Africana/tratamento farmacológico , Tripanossomíase Africana/parasitologia
14.
Mol Biol Cell ; 29(9): 1100-1110, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29496964

RESUMO

Components of the nuclear periphery coordinate a multitude of activities, including macromolecular transport, cell-cycle progression, and chromatin organization. Nuclear pore complexes (NPCs) mediate nucleocytoplasmic transport, mRNA processing, and transcriptional regulation, and NPC components can define regions of high transcriptional activity in some organisms at the nuclear periphery and nucleoplasm. Lineage-specific features underpin several core nuclear functions and in trypanosomatids, which branched very early from other eukaryotes, unique protein components constitute the lamina, kinetochores, and parts of the NPCs. Here we describe a phenylalanine-glycine (FG)-repeat nucleoporin, TbNup53b, that has dual localizations within the nucleoplasm and NPC. In addition to association with nucleoporins, TbNup53b interacts with a known trans-splicing component, TSR1, and has a role in controlling expression of surface proteins including the nucleolar periphery-located, procyclin genes. Significantly, while several nucleoporins are implicated in intranuclear transcriptional regulation in metazoa, TbNup53b appears orthologous to components of the yeast/human Nup49/Nup58 complex, for which no transcriptional functions are known. These data suggest that FG-Nups are frequently co-opted to transcriptional functions during evolution and extend the presence of FG-repeat nucleoporin control of gene expression to trypanosomes, suggesting that this is a widespread and ancient eukaryotic feature, as well as underscoring once more flexibility within nucleoporin function.


Assuntos
Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Transporte Ativo do Núcleo Celular , Antígenos de Superfície/imunologia , Núcleo Celular/metabolismo , Sequência Conservada , Glicina , Poro Nuclear/metabolismo , Fenilalanina , Domínios Proteicos , Elementos Estruturais de Proteínas , Alinhamento de Sequência , Trypanosoma/metabolismo , Trypanosoma brucei brucei/metabolismo
15.
Trends Parasitol ; 34(3): 197-207, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29396200

RESUMO

Trypanosoma brucei gambiense causes human African trypanosomiasis (HAT). Between 1990 and 2015, almost 440000 cases were reported. Large-scale screening of populations at risk, drug donations, and efforts by national and international stakeholders have brought the epidemic under control with <2200 cases in 2016. The World Health Organization (WHO) has set the goals of gambiense-HAT elimination as a public health problem for 2020, and of interruption of transmission to humans for 2030. Latent human infections and possible animal reservoirs may challenge these goals. It remains largely unknown whether, and to what extend, they have an impact on gambiense-HAT transmission. We argue that a better understanding of the contribution of human and putative animal reservoirs to gambiense-HAT epidemiology is mandatory to inform elimination strategies.


Assuntos
Erradicação de Doenças , Reservatórios de Doenças , Tripanossomíase Africana/prevenção & controle , Tripanossomíase Africana/transmissão , Animais , Humanos , Fatores de Risco , Trypanosoma brucei gambiense/fisiologia , Tripanossomíase Africana/epidemiologia , Tripanossomíase Africana/parasitologia
16.
Cell Rep ; 17(3): 660-670, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27732844

RESUMO

During infection in mammals, the protozoan parasite Trypanosoma brucei transforms from a proliferative bloodstream form to a quiescent form that is pre-adapted to host transition. AMP analogs are known to induce quiescence and also inhibit TbTOR4. To examine the role of AMP-activated kinase (AMPK) in the regulation of this developmental transition, we characterized trypanosome TbAMPK complexes. Expression of a constitutively active AMPKα1 induces quiescence of the infective form, and TbAMPKα1 phosphorylation occurs during differentiation of wild-type pleomorphic trypanosomes to the quiescent stumpy form in vivo. Compound C, a well-known AMPK inhibitor, inhibits parasite differentiation in mice. We also provide evidence linking oxidative stress to TbAMPKα1 activation and quiescent differentiation, suggesting that TbAMPKα1 activation balances quiescence, proliferation, and differentiation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Ciclo Celular , Transdução de Sinais , Trypanosoma brucei brucei/citologia , Trypanosoma brucei brucei/enzimologia , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Monofosfato de Adenosina/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Tripanossomíase/parasitologia , Tripanossomíase/patologia
17.
Nucleic Acids Res ; 44(22): 10554-10570, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27625397

RESUMO

The nuclear lamina is a filamentous structure subtending the nuclear envelope and required for chromatin organization, transcriptional regulation and maintaining nuclear structure. The trypanosomatid coiled-coil NUP-1 protein is a lamina component functionally analogous to lamins, the major lamina proteins of metazoa. There is little evidence for shared ancestry, suggesting the presence of a distinct lamina system in trypanosomes. To find additional trypanosomatid lamina components we identified NUP-1 interacting proteins by affinity capture and mass-spectrometry. Multiple components of the nuclear pore complex (NPC) and a second coiled-coil protein, which we termed NUP-2, were found. NUP-2 has a punctate distribution at the nuclear periphery throughout the cell cycle and is in close proximity to NUP-1, the NPCs and telomeric chromosomal regions. RNAi-mediated silencing of NUP-2 leads to severe proliferation defects, gross alterations to nuclear structure, chromosomal organization and nuclear envelope architecture. Further, transcription is altered at telomere-proximal variant surface glycoprotein (VSG) expression sites (ESs), suggesting a role in controlling ES expression, although NUP-2 silencing does not increase VSG switching. Transcriptome analysis suggests specific alterations to Pol I-dependent transcription. NUP-1 is mislocalized in NUP-2 knockdown cells and vice versa, implying that NUP-1 and NUP-2 form a co-dependent network and identifying NUP-2 as a second trypanosomatid nuclear lamina component.


Assuntos
Lâmina Nuclear/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas de Protozoários/metabolismo , Trypanosoma brucei brucei/metabolismo , Cromossomos/genética , Cromossomos/metabolismo , Dano ao DNA , Regulação da Expressão Gênica , Lâmina Nuclear/ultraestrutura , Poro Nuclear/metabolismo , Poro Nuclear/ultraestrutura , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Transporte Proteico , Proteínas de Protozoários/genética , Transcriptoma , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/ultraestrutura
18.
Front Microbiol ; 6: 765, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26257727

RESUMO

Gambiense trypanosomiasis is considered an anthroponotic disease. Consequently, control programs are generally aimed at stopping transmission of Trypanosoma brucei gambiense (T. b. gambiense) by detecting and treating human cases. However, the persistence of numerous foci despite efforts to eliminate this disease questions this strategy as unique tool to pursue the eradication. The role of animals as a reservoir of T. b. gambiense is still controversial, but could partly explain maintenance of the infection at hypo-endemic levels. In the present study, we evaluated the presence of T. b. gambiense in wild animals in Equatorial Guinea. The infection rate ranged from 0.8% in the insular focus of Luba to more than 12% in Mbini, a focus with a constant trickle of human cases. The parasite was detected in a wide range of animal species including four species never described previously as putative reservoirs. Our study comes to reinforce the hypothesis that animals may play a role in the persistence of T. b. gambiense transmission, being particularly relevant in low transmission settings. Under these conditions the integration of sustained vector control and medical interventions should be considered to achieve the elimination of gambiense trypanosomiasis.

19.
Cell Microbiol ; 17(10): 1523-35, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25924022

RESUMO

African trypanosomes infect a broad range of mammals, but humans and some higher primates are protected by serum trypanosome lytic factors that contain apolipoprotein L1 (ApoL1). In the human-infective subspecies of Trypanosoma brucei, Trypanosoma brucei rhodesiense, a gene product derived from the variant surface glycoprotein gene family member, serum resistance-associated protein (SRA protein), protects against ApoL1-mediated lysis. Protection against trypanosome lytic factor requires the direct interaction between SRA protein and ApoL1 within the endocytic apparatus of the trypanosome, but some uncertainty remains as to the precise mechanism and location of this interaction. In order to provide more insight into the mechanism of SRA-mediated resistance to trypanosome lytic factor, we assessed the localization of SRA in T. b. rhodesiense EATRO3 using a novel monoclonal antibody raised against SRA together with a set of well-characterized endosomal markers. By three-dimensional deconvolved immunofluorescence single-cell analysis, combined with double-labelling immunoelectron microscopy, we found that ≈ 50% of SRA protein localized to the lysosome, with the remaining population being distributed through the endocytic pathway, but apparently absent from the flagellar pocket membrane. These data suggest that the SRA/trypanolytic factor interaction is intracellular, with the concentration within the endosomes potentially crucial for ensuring a high efficiency.


Assuntos
Endossomos/química , Lisossomos/química , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/genética , Proteínas de Protozoários/análise , Proteínas de Protozoários/genética , Trypanosoma brucei brucei/química , Trypanosoma brucei rhodesiense/química , Animais , Apolipoproteína L1 , Apolipoproteínas/metabolismo , Humanos , Lipoproteínas HDL/metabolismo , Microscopia de Fluorescência , Microscopia Imunoeletrônica , Trypanosoma brucei brucei/efeitos dos fármacos , Trypanosoma brucei brucei/imunologia , Trypanosoma brucei rhodesiense/efeitos dos fármacos , Trypanosoma brucei rhodesiense/imunologia
20.
PLoS Pathog ; 10(12): e1004545, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25474309

RESUMO

Bloodstream form trypanosomes avoid the host immune response by switching the expression of their surface proteins between Variant Surface Glycoproteins (VSG), only one of which is expressed at any given time. Monoallelic transcription of the telomeric VSG Expression Site (ES) by RNA polymerase I (RNA pol I) localizes to a unique nuclear body named the ESB. Most work has focused on silencing mechanisms of inactive VSG-ESs, but the mechanisms involved in transcriptional activation of a single VSG-ES remain largely unknown. Here, we identify a highly SUMOylated focus (HSF) in the nucleus of the bloodstream form that partially colocalizes with the ESB and the active VSG-ES locus. SUMOylation of chromatin-associated proteins was enriched along the active VSG-ES transcriptional unit, in contrast to silent VSG-ES or rDNA, suggesting that it is a distinct feature of VSG-ES monoallelic expression. In addition, sequences upstream of the active VSG-ES promoter were highly enriched in SUMOylated proteins. We identified TbSIZ1/PIAS1 as the SUMO E3 ligase responsible for SUMOylation in the active VSG-ES chromatin. Reduction of SUMO-conjugated proteins by TbSIZ1 knockdown decreased the recruitment of RNA pol I to the VSG-ES and the VSG-ES-derived transcripts. Furthermore, cells depleted of SUMO conjugated proteins by TbUBC9 and TbSUMO knockdown confirmed the positive function of SUMO for VSG-ES expression. In addition, the largest subunit of RNA pol I TbRPA1 was SUMOylated in a TbSIZ-dependent manner. Our results show a positive mechanism associated with active VSG-ES expression via post-translational modification, and indicate that chromatin SUMOylation plays an important role in the regulation of VSG-ES. Thus, protein SUMOylation is linked to active gene expression in this protozoan parasite that diverged early in evolution.


Assuntos
Glicoproteínas/biossíntese , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas de Protozoários/metabolismo , Sumoilação/fisiologia , Trypanosoma brucei brucei/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Regulação da Expressão Gênica/fisiologia , Glicoproteínas/genética , Proteínas Inibidoras de STAT Ativados/genética , Processamento de Proteína Pós-Traducional/fisiologia , Proteínas de Protozoários/genética , Trypanosoma brucei brucei/genética , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...