Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 85
1.
J Epidemiol Glob Health ; 10(4): 280-287, 2020 12.
Article En | MEDLINE | ID: mdl-32959623

Trypanosoma brucei rhodesiense Human African Trypanosomiasis (rHAT) is a zoonotic disease transmitted by tsetse flies from wild and domestic animals. It presents as an acute disease and advances rapidly into a neurological form that can only be treated with melarsoprol, which is associated with a high fatality rate. Bringing diagnostic services for rHAT closer to at-risk populations would increase chances of detecting cases in early stages of disease when treatment is safer and more effective. In Malawi, most of the rHAT cases occur around Vwaza Marsh Wildlife Reserve. Until 2013, diagnosis of rHAT in the region was only available at the Rumphi District Hospital that is more than 60 km away from the reserve. In 2013, Malawi's Ministry of Health initiated a project to enhance the detection of rHAT in five health facilities around Vwaza Marsh by upgrading laboratories and training technicians. We report here a retrospective study that was carried out to evaluate the impact of improving access to diagnostic services on the disease stage at diagnosis and on mortality. Between August 2014 and July 2017, 2014 patients suspected of having the disease were tested by microscopy, including 1267 who were tested in the new facilities. This resulted in the identification of 78 new rHAT cases, of which six died. Compared with previous years, data obtained during this period indicate that access to diagnostic services closer to where people at the greatest risk of infection live promotes identification of cases in earlier stages of infection, and improves treatment outcomes.


Health Services Accessibility , Trypanosoma brucei rhodesiense , Trypanosomiasis, African , Animals , Early Diagnosis , Health Services Accessibility/statistics & numerical data , Humans , Malawi/epidemiology , Retrospective Studies , Trypanosoma brucei rhodesiense/isolation & purification , Trypanosomiasis, African/diagnosis , Trypanosomiasis, African/mortality , Zoonoses
2.
Proc Natl Acad Sci U S A ; 116(48): 24214-24220, 2019 11 26.
Article En | MEDLINE | ID: mdl-31723045

Although CRIg was originally identified as a macrophage receptor for binding complement C3b/iC3b in vitro, recent studies reveal that CRIg functions as a pattern recognition receptor in vivo for Kupffer cells (KCs) to directly bind bacterial pathogens in a complement-independent manner. This raises the critical question of whether CRIg captures circulating pathogens through interactions with complement in vivo under flow conditions. Furthermore, the role of CRIg during parasitic infection is unknown. Taking advantage of intravital microscopy and using African trypanosomes as a model, we studied the role of CRIg in intravascular clearance of bloodborne parasites. Complement C3 is required for intravascular clearance of African trypanosomes by KCs, preventing the early mortality of infected mice. Moreover, antibodies are essential for complement-mediated capture of circulating parasites by KCs. Interestingly, reduced antibody production was observed in the absence of complement C3 during infection. We further demonstrate that CRIg but not CR3 is critically involved in KC-mediated capture of circulating parasites, accounting for parasitemia control and host survival. Of note, CRIg cannot directly catch circulating parasites and antibody-induced complement activation is indispensable for CRIg-mediated parasite capture. Thus, we provide evidence that CRIg, by interacting with complement in vivo, plays an essential role in intravascular clearance of bloodborne parasites. Targeting CRIg may be considered as a therapeutic strategy.


Complement C3b/metabolism , Host-Parasite Interactions/physiology , Parasitemia/parasitology , Receptors, Complement/physiology , Trypanosomiasis, African/blood , Animals , Complement C3b/immunology , Intravital Microscopy , Kupffer Cells/immunology , Kupffer Cells/parasitology , Macrophage-1 Antigen/metabolism , Macrophages/parasitology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/pathogenicity , Trypanosoma congolense/pathogenicity , Trypanosomiasis, African/mortality , Trypanosomiasis, African/parasitology
3.
Exp Parasitol ; 205: 107714, 2019 Oct.
Article En | MEDLINE | ID: mdl-31279927

The objective of the present study was to evaluate the clinical signs, electrocardiographic signs and evolution of histopathological lesions in the heart of sheep experimentally infected by Trypanosoma vivax during the acute and chronic phases of infection as well as to investigate the presence of parasitic DNA in the heart using polymerase chain reaction (PCR). Twenty-two male sheep were divided into the following four groups: G1, which consisted of six sheep infected by T. vivax that were evaluated until 20 days post-infection (dpi; acute phase); G2, which consisted of six sheep infected by T. vivax that were evaluated until 90 dpi (chronic phase); and G3 and G4 groups, which each consisted of five uninfected sheep. At the end of the experimental period, electrocardiographic evaluations and necroscopic examinations were performed. Fragments of the heart were collected and stained by Hematoxylin-Eosin and Masson's trichrome, and the fragments were also evaluated by PCR for T. vivax. G2 animals presented clinical signs suggestive of heart failure and electrocardiogram alterations characterized by prolonged P, T and QRS complex durations as well as by a cardiac electrical axis shift to the left and increased heart rate. In these animals, mononuclear multifocal myocarditis and interstitial fibrosis were also observed. PCR revealed positivity for T. vivax in two G1 animals and in all G2 animals. Thus, these findings suggested that T. vivax is responsible for the occurrence of cardiac lesions, which are related to heart failure, electrocardiographic alterations and mortality of the infected animals.


DNA, Protozoan/isolation & purification , Heart Failure/veterinary , Heart/parasitology , Sheep Diseases/parasitology , Trypanosoma vivax/pathogenicity , Trypanosomiasis, African/veterinary , Acute Disease , Animals , Antibodies, Protozoan/blood , Chronic Disease/veterinary , Electrocardiography/veterinary , Fluorescent Antibody Technique, Indirect/veterinary , Heart Failure/mortality , Heart Failure/parasitology , Immunoglobulin G/blood , Male , Myocardium/pathology , Parasitemia/veterinary , Pericarditis/parasitology , Pericarditis/pathology , Pericarditis/veterinary , Polymerase Chain Reaction/veterinary , Random Allocation , Sheep , Sheep Diseases/mortality , Sheep Diseases/pathology , Trypanosoma vivax/genetics , Trypanosoma vivax/immunology , Trypanosoma vivax/isolation & purification , Trypanosomiasis, African/complications , Trypanosomiasis, African/mortality , Trypanosomiasis, African/pathology
4.
Exp Parasitol ; 199: 40-46, 2019 Apr.
Article En | MEDLINE | ID: mdl-30840850

Human African trypanosomosis (HAT) and animal African trypanosomosis (AAT) are diseases of economic importance in humans and animals that affect more than 36 African countries. The currently available trypanocidal drugs are associated with side effects, and the parasites are continually developing resistance. Thus, effective and safe drugs are needed for the treatment of HAT and AAT. This study aimed to evaluate the effects of azithromycin (AZM) on Trypanosoma brucei brucei-infected mice. Mice were randomly divided into 7 groups consisting of a vehicle control group, 5 test groups and a diminazene aceturate (DA)-treated group. Mice were treated orally for 7 and 28 days, as short-term and long-term treatments, respectively. Short-term AZM treatment cured 23% (16 of 70) of the overall treated mice whereas long-term treatment resulted in the survival of 70% of the mice in the groups that received AZM at doses of 300 and 400 mg/kg. Trypanosomes treated in vitro with 25 µg/mL of AZM were subjected to transmission electron microscopy, which revealed the presence of increased numbers of glycosomes and acidocalcisomes in comparison to the vehicle group. The current study showed the trypanocidal effect of AZM on T. b. brucei in vivo. The demonstrated efficacy increased with an increase in treatment period and an increased concentration of AZM.


Anti-Infective Agents/administration & dosage , Azithromycin/administration & dosage , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/drug therapy , Administration, Oral , Animals , Anti-Infective Agents/pharmacology , Anti-Infective Agents/therapeutic use , Azithromycin/pharmacology , Azithromycin/therapeutic use , Body Weight/drug effects , Female , Inhibitory Concentration 50 , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Parasitemia/drug therapy , Random Allocation , Survival Rate , Time Factors , Trypanosoma brucei brucei/ultrastructure , Trypanosomiasis, African/mortality
5.
PLoS Negl Trop Dis ; 12(6): e0006504, 2018 06.
Article En | MEDLINE | ID: mdl-29897919

We conducted a retrospective study on mortality trends and risk factors in 781 naïve cases of advanced stage-2 sleeping sickness admitted between 1989 and 2012 at the National Reference Center for Human African Trypanosomiasis (HAT), Department of Neurology, Kinshasa University, Democratic Republic of Congo (DRC). Death was the outcome variable whereas age, gender, duration of disease, location of trypanosomes in body fluids, cytorachy, protidorachy, clinical status (assessed on a syndromic and functional basis) on admission, and treatment regimen were predictors in logistic regression models run at the 0.05 significance level. Death proportions were 17.2% in the standard melarsoprol schedule (3-series of intravenous melarsoprol on 3 successive days at 3.6 mg/kg/d, with a one-week interval between the series, ARS 9); 12.1% in the short schedule melarsoprol (10 consecutive days of intravenous melarsoprol at 2.2 mg/kg/d, ARS 10), 5.4% in the first-line eflornithine (14 days of eflornithine at 400 mg/kg/d in 4 infusions a day DFMO B), 9.1% in the NECT treatment regimen (eflornithine for 7 days at 400, mg/kg/d in 2 infusions a day combined with oral nifurtimox for 10 days at 15 mg/kg/d in 3 doses a day); and high (36%) in the group with select severely affected patients given eflornithine because of their clinical status on admission, at the time when this expensive drug was kept for treatment of relapses (14 days at 400 mg/kg/d in 4 infusions a day, DFMO A). After adjusting for treatment, death odds ratios were as follows: 10.40 [(95% CI: 6.55-16.51); p = .000] for clinical dysfunction (severely impaired clinical status) on admission, 2.14 [(95% CI: 1.35-3.39); p = .001] for high protidorachy, 1.99 [(95% CI: 1.18-3.37); p = .010] for the presence of parasites in the CSF and 1.70 [(95% CI: 1.03-2.81); p = .038] for high cytorachy. A multivariable analysis within treatment groups retained clinical status on admission (in ARS 9, ARS 10 and DFMO B groups) and high protidorachy (in ARS 10 and DFMO B groups) as significant predictors of death. The algorithm for initial clinical status assessment used in the present study may serve as the basis for further development of standardized assessment tools relevant to the clinical management of HAT and information exchange in epidemiological reports.


Trypanosomiasis, African/epidemiology , Trypanosomiasis, African/mortality , Adolescent , Adult , Democratic Republic of the Congo/epidemiology , Disease Management , Drug Therapy, Combination , Eflornithine/administration & dosage , Eflornithine/therapeutic use , Female , Hospital Records , Humans , Male , Melarsoprol/administration & dosage , Melarsoprol/therapeutic use , Middle Aged , Multivariate Analysis , Nifurtimox/administration & dosage , Nifurtimox/therapeutic use , Recurrence , Retrospective Studies , Risk Factors , Treatment Outcome , Trypanocidal Agents/administration & dosage , Trypanocidal Agents/therapeutic use , Trypanosoma brucei gambiense/drug effects , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/parasitology , Young Adult
6.
Infect Genet Evol ; 63: 269-276, 2018 09.
Article En | MEDLINE | ID: mdl-29807131

Trypanosoma brucei gambiense (T. b. gambiense) is the major causative agent of human African trypanosomiasis (HAT). A great variety of clinical outcomes have been observed in West African foci, probably due to complex host-parasite interactions. In order to separate the roles of parasite genetic diversity and host variability, we have chosen to precisely characterize the pathogenicity and virulence of T. b. gambiense field isolates in a mouse model. Thirteen T. b. gambiense strains were studied in experimental infections, with 20 Balb/C infected mice per isolate. Mice were monitored for 30 days, in which mortality, parasitemia, anemia, and weight were recorded. Mortality rate, prepatent period, and maximum parasitemia were estimated, and a survival analysis was performed to compare strain pathogenicity. Mixed models were used to assess parasitemia dynamics, weight, and changes in Packed Cell Volume (PCV). Finally, a multivariate analysis was performed to infer relationships between all variables. A large phenotypic diversity was observed. Pathogenicity was highly variable, ranging from strains that kill their host within 9 days to a non-pathogenic strain (no deaths during the experiment). Virulence was also variable, with maximum parasitemia values ranging from 42 million to 1 billion trypanosomes/ml. Reduced PCV and weight occurred in the first two weeks of the infection, with the exception of two strains. Finally, the global analysis highlighted three groups of strains: a first group with highly pathogenic strains showing an early mortality associated with a short prepatent period; a second group of highly virulent strains with intermediate pathogenicity; and a third group of isolates characterized by low pathogenicity and virulence patterns. Such biological differences could be related to the observed clinical diversity in HAT. A better understanding of the biological pathways underlying the observed phenotypic diversity could thus help to clarify the complex nature of the host-parasite interactions that determine the resistance/susceptibility status to T. brucei gambiense.


Host-Parasite Interactions , Parasitemia/pathology , Phenotype , Trypanosoma brucei gambiense/pathogenicity , Trypanosomiasis, African/pathology , Africa, Western , Animals , Body Weight , Disease Models, Animal , Erythrocyte Indices , Erythrocytes/parasitology , Erythrocytes/pathology , Humans , Mice , Mice, Inbred BALB C , Multivariate Analysis , Parasitemia/mortality , Parasitemia/parasitology , Principal Component Analysis , Survival Analysis , Trypanosoma brucei gambiense/classification , Trypanosoma brucei gambiense/isolation & purification , Trypanosomiasis, African/mortality , Trypanosomiasis, African/parasitology , Virulence
7.
PLoS Negl Trop Dis ; 12(2): e0006188, 2018 02.
Article En | MEDLINE | ID: mdl-29425200

BACKGROUND: This paper presents the development of an agent-based model (ABM) to incorporate climatic drivers which affect tsetse fly (G. m. morsitans) population dynamics, and ultimately disease transmission. The model was used to gain a greater understanding of how tsetse populations fluctuate seasonally, and investigate any response observed in Trypanosoma brucei rhodesiense human African trypanosomiasis (rHAT) disease transmission, with a view to gaining a greater understanding of disease dynamics. Such an understanding is essential for the development of appropriate, well-targeted mitigation strategies in the future. METHODS: The ABM was developed to model rHAT incidence at a fine spatial scale along a 75 km transect in the Luangwa Valley, Zambia. The model incorporates climatic factors that affect pupal mortality, pupal development, birth rate, and death rate. In combination with fine scale demographic data such as ethnicity, age and gender for the human population in the region, as well as an animal census and a sample of daily routines, we create a detailed, plausible simulation model to explore tsetse population and disease transmission dynamics. RESULTS: The seasonally-driven model suggests that the number of infections reported annually in the simulation is likely to be a reasonable representation of reality, taking into account the high levels of under-detection observed. Similar infection rates were observed in human (0.355 per 1000 person-years (SE = 0.013)), and cattle (0.281 per 1000 cattle-years (SE = 0.025)) populations, likely due to the sparsity of cattle close to the tsetse interface. The model suggests that immigrant tribes and school children are at greatest risk of infection, a result that derives from the bottom-up nature of the ABM and conditioning on multiple constraints. This result could not be inferred using alternative population-level modelling approaches. CONCLUSIONS: In producing a model which models the tsetse population at a very fine resolution, we were able to analyse and evaluate specific elements of the output, such as pupal development and the progression of the teneral population, allowing the development of our understanding of the tsetse population as a whole. This is an important step in the production of a more accurate transmission model for rHAT which can, in turn, help us to gain a greater understanding of the transmission system as a whole.


Climate , Seasons , Trypanosoma brucei rhodesiense/pathogenicity , Trypanosomiasis, African/epidemiology , Trypanosomiasis, African/transmission , Tsetse Flies/physiology , Adolescent , Adult , Age Factors , Animals , Birth Rate , Cattle , Child , Child, Preschool , Ethnicity , Female , Geographic Mapping , Humans , Incidence , Infant , Male , Middle Aged , Mortality , Population Dynamics , Rain , Schools , Sex Factors , Temperature , Trypanosomiasis, African/mortality , Young Adult , Zambia
8.
Parasitology ; 145(9): 1235-1242, 2018 08.
Article En | MEDLINE | ID: mdl-29362015

This study assessed the virulence of Trypanosoma evansi, the causative agent of camel trypanosomiasis (surra), affecting mainly camels among other hosts in Africa, Asia and South America, with high mortality and morbidity. Using Swiss white mice, we assessed virulence of 17 T. evansi isolates collected from surra endemic countries. We determined parasitaemia, live body weight, packed cell volume (PCV) and survivorship in mice, for a period of 60 days' post infection. Based on survivorship, the 17 isolates were classified into three virulence categories; low (31-60 days), moderate (11-30 days) and high (0-10 days). Differences in survivorship, PCV and bodyweights between categories were significant and correlated (P < 0.05). Of the 10 Kenyan isolates, four were of low, five moderate and one (Type B) of high virulence. These findings suggest differential virulence between T. evansi isolates. In conclusion, these results show that the virulence of T. evansi may be region specific, the phenotype of the circulating parasite should be considered in the management of surra. There is also need to collect more isolates from other surra endemic regions to confirm this observation.


Parasitemia/veterinary , Trypanosoma/pathogenicity , Trypanosomiasis, African/mortality , Animals , Camelus/parasitology , Mice , Trypanosoma/genetics , Virulence
9.
Lancet ; 391(10116): 144-154, 2018 01 13.
Article En | MEDLINE | ID: mdl-29113731

BACKGROUND: Few therapeutic options are available to treat the late-stage of human African trypanosomiasis, a neglected tropical disease, caused by Trypanosoma brucei gambiense (g-HAT). The firstline treatment is a combination therapy of oral nifurtimox and intravenous eflornithine that needs to be administered in a hospital setting by trained personnel, which is not optimal given that patients often live in remote areas with few health resources. Therefore, we aimed to assess the safety and efficacy of an oral regimen of fexinidazole (a 2-substituted 5-nitroimidazole with proven trypanocidal activity) versus nifurtimox eflornithine combination therapy in patients with late-stage g-HAT. METHODS: In this randomised, phase 2/3, open-label, non-inferiority trial, we recruited patients aged 15 years and older with late-stage g-HAT from g-HAT treatment centres in the Democratic Republic of the Congo (n=9) and the Central African Republic (n=1). Patients were randomly assigned (2:1) to receive either fexinidazole or nifurtimox eflornithine combination therapy according to a predefined randomisation list (block size six). The funder, data management personnel, and study statisticians were masked to treatment. Oral fexinidazole was given once a day (days 1-4: 1800 mg, days 5-10: 1200 mg). Oral nifurtimox was given three times a day (days 1-10: 15 mg/kg per day) with eflornithine twice a day as 2 h infusions (days 1-7: 400 mg/kg per day). The primary endpoint was success at 18 months (ie, deemed as patients being alive, having no evidence of trypanosomes in any body fluid, not requiring rescue medication, and having a cerebrospinal fluid white blood cell count ≤20 cells per µL). Safety was assessed through routine monitoring. Primary efficacy analysis was done in the modified intention-to-treat population and safety analyses in the intention-to-treat population. The acceptable margin for the difference in success rates was defined as 13%. This study has been completed and is registered with ClinicalTrials.gov, number NCT01685827. FINDINGS: Between October, 2012, and November, 2016, 419 patients were pre-screened. Of the 409 eligible patients, 14 were not included because they did not meet all inclusion criteria (n=12) or for another reason (n=2). Therefore, 394 patients were randomly assigned, 264 to receive fexinidazole and 130 to receive nifurtimox eflornithine combination therapy. Success at 18 months was recorded in 239 (91%) patients given fexinidazole and 124 (98%) patients given nifurtimox eflornithine combination therapy, within the margin of acceptable difference of -6·4% (97·06% CI -11·2 to -1·6; p=0·0029). We noted no difference in the proportion of patients who experienced treatment-related adverse events (215 [81%] in the fexinidazole group vs 102 [79%] in the nifurtimox eflornithine combination therapy group). Treatment discontinuations were unrelated to treatment (n=2 [1%] in the fexinidazole group). Temporary nifurtimox eflornithine combination therapy interruption occurred in three (2%) patients. 11 patients died during the study (nine [3%] in the fexinidazole group vs two [2%] in the nifurtimox eflornithine combination therapy group). INTERPRETATION: Our findings show that oral fexinidazole is effective and safe for the treatment of T b gambiense infection compared with nifurtimox eflornithine combination therapy in late-stage HAT patients. Fexinidazole could be a key asset in the elimination of this fatal neglected disease. FUNDING: Drugs for Neglected Diseases initiative.


Nifurtimox/therapeutic use , Nitroimidazoles/therapeutic use , Trypanocidal Agents/therapeutic use , Trypanosoma brucei gambiense , Trypanosomiasis, African/drug therapy , Administration, Oral , Adult , Democratic Republic of the Congo , Drug Administration Schedule , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Treatment Outcome , Trypanosomiasis, African/diagnosis , Trypanosomiasis, African/mortality
10.
Antimicrob Agents Chemother ; 60(4): 2532-6, 2016 Apr.
Article En | MEDLINE | ID: mdl-26787703

Current treatments for African trypanosomiasis are either toxic, costly, difficult to administer, or prone to elicit resistance. This study evaluated the activity of bisnaphthalimidopropyl (BNIP) derivatives againstTrypanosoma brucei BNIPDiaminobutane (BNIPDabut), the most active of these compounds, showedin vitroinhibition in the single-unit nanomolar range, similar to the activity in the reference drug pentamidine, and presented low toxicity and adequate metabolic stability. Additionally, using a murine model of acute infection and live imaging, a significant decrease in parasite load in BNIPDabut-treated mice was observed. However, cure was not achieved. BNIPDabut constitutes a new scaffold for antitrypanosomal drugs that deserves further consideration.


Naphthalimides/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/drug therapy , Animals , Cell Line , Drug Stability , Female , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inhibitory Concentration 50 , Liver/drug effects , Liver/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Naphthalimides/chemical synthesis , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Parasite Load , Pentamidine/pharmacology , Primary Cell Culture , Structure-Activity Relationship , Survival Analysis , Trypanocidal Agents/chemical synthesis , Trypanosoma brucei brucei/growth & development , Trypanosoma brucei brucei/metabolism , Trypanosomiasis, African/mortality , Trypanosomiasis, African/parasitology , Trypanosomiasis, African/pathology
11.
Parasit Vectors ; 8: 564, 2015 Oct 28.
Article En | MEDLINE | ID: mdl-26510460

BACKGROUND: The Brazilian Semiarid is the home of the largest herd of donkeys in South America and of outbreaks of Trypanosoma vivax infection of high mortality in dairy cattle and sheep. For a comprehensive understanding of the underlying mechanisms of these outbreaks and epidemiological role of donkeys, we surveyed for T. vivax in wandering donkeys and follow the experimental infection of donkeys and sheep with a highly virulent isolate from the Semiarid. METHODS: Blood samples from 180 randomly selected wandering donkeys from the Brazilian Semiarid region were employed for PCV and parasitemia assessments and tested using the T. vivax-specific TviCATL-PCR assay. PCR-amplifed Cathepsin L (CATL) sequences were employed for genotyping and phylogenetic analysis. Four wandering donkeys were experimentally infected with a T. vivax isolate obtained during an outbreak of high mortality in the Semiarid; the control group consisted of two non-inoculated donkeys. RESULTS: We detected T. vivax in 30 of 180 wandering donkeys (16.6 %) using TviCATL-PCR. The prevalence was higher during the dry (15.5 %) than the wet season (1.1 %) and more females (23.1 %) than males (8.9 %) were infected. All the PCR-positive donkeys lacked patent parasitemia and showed normal values of body condition score (BCS) and packed cell volume (PCV). To evaluate the probable tolerance of donkeys to T. vivax, we inoculated five donkeys with a highly virulent isolate (TviBrRp) from the Semiarid. All inoculated donkeys became PCR-positive, but their parasitemia was always subpatent. A control goat inoculated with TviBrRp showed increasing parasitemia concurrently with fever, declining PCV, tachycardia, mucous membrane pallor, enlarged lymph nodes and anorexia. None of these signs were observed in donkeys. However, T. vivax from wandering donkeys shared identical or highly similar genotypes (identified by Cathepsin L sequences) with isolates from cattle and sheep outbreaks of acute disease in the Semiarid. CONCLUSIONS: This is the first report of T. vivax in donkeys in Brazil and, to our knowledge, the first experimental infection of donkeys with T. vivax. The symptomless field and experimental infections corroborated that donkeys are more tolerant to T. vivax than other livestock species as shown in African countries. Therefore, farmers, veterinaries and control programmes should be aware of healthy carrier donkeys as a possible source of T. vivax for susceptible livestock species in the Brazilian Semiarid.


Cattle Diseases/epidemiology , Disease Outbreaks/veterinary , Equidae/parasitology , Sheep Diseases/epidemiology , Trypanosoma vivax/isolation & purification , Trypanosomiasis, African/veterinary , Animals , Brazil/epidemiology , Carrier State , Cattle , Cattle Diseases/mortality , Female , Goats , Livestock , Male , Parasitemia/veterinary , Prevalence , Sheep , Sheep Diseases/mortality , Trypanosoma vivax/genetics , Trypanosomiasis, African/epidemiology , Trypanosomiasis, African/mortality
12.
Antimicrob Agents Chemother ; 59(12): 7530-9, 2015 Dec.
Article En | MEDLINE | ID: mdl-26392508

We report the results of a screen of a library of 925 potential prenyl synthase inhibitors against Trypanosoma brucei farnesyl diphosphate synthase (TbFPPS) and against T. brucei, the causative agent of human African trypanosomiasis. The most potent compounds were lipophilic analogs of the bone resorption drug zoledronate, some of which had submicromolar to low micromolar activity against bloodstream form T. brucei and selectivity indices of up to ∼ 300. We evaluated the effects of two such inhibitors on survival and parasitemia in a T. brucei mouse model of infection and found that survival increased by up to 16 days. We also investigated the binding of three lipophilic bisphosphonates to an expressed TbFPPS using crystallography and investigated the thermodynamics of binding using isothermal titration calorimetry.


Diphosphonates/pharmacology , Enzyme Inhibitors/pharmacology , Geranyltranstransferase/chemistry , Parasitemia/drug therapy , Protozoan Proteins/chemistry , Trypanocidal Agents/pharmacology , Trypanosomiasis, African/drug therapy , Animals , Binding Sites , Diphosphonates/chemical synthesis , Diphosphonates/chemistry , Disease Models, Animal , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Geranyltranstransferase/antagonists & inhibitors , Geranyltranstransferase/genetics , Geranyltranstransferase/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Inbred BALB C , Models, Molecular , Parasitemia/mortality , Parasitemia/parasitology , Parasitemia/pathology , Protein Binding , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Survival Analysis , Thermodynamics , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/chemistry , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/growth & development , Trypanosomiasis, African/mortality , Trypanosomiasis, African/parasitology , Trypanosomiasis, African/pathology
13.
Parasit Vectors ; 7: 210, 2014 May 03.
Article En | MEDLINE | ID: mdl-24885708

BACKGROUND: Mechanical transmission of the major livestock pathogen Trypanosoma vivax by other biting flies than tsetse allows its spread from Africa to the New World. Genetic studies are restricted to a small number of isolates and based on molecular markers that evolve too slowly to resolve the relationships between American and West African populations and, thus, unable us to uncover the recent history of T. vivax in the New World. METHODS: T. vivax genetic diversity, population structure and the source of outbreaks was investigated through the microsatellite multiloci (7 loci) genotype (MLGs) analysis in South America (47isolates from Brazil, Venezuela and French Guiana) and West Africa (12 isolates from The Gambia, Burkina Faso, Ghana, Benin and Nigeria). Relationships among MLGs were explored using phylogenetic, principal component and STRUCTURE analyses. RESULTS: Although closely phylogenetically related, for the first time, genetic differences were detected between T. vivax isolates from South America (11 genotypes/47 isolates) and West Africa (12 genotypes/12 isolates) with no MLGs in common. Diversity was far greater across West Africa than in South America, where genotypes from Brazil (MLG1-6), Venezuela (MLG7-10) and French Guiana (MLG11) shared similar but not identical allele composition. No MLG was exclusive to asymptomatic (endemic areas) or sick (outbreaks in non-endemic areas) animals, but only MLGs1, 2 and 3 were responsible for severe haematological and neurological disorders. CONCLUSIONS: Our results revealed closely related genotypes of T. vivax in Brazil and Venezuela, regardless of endemicity and clinical conditions of the infected livestock. The MLGs analysis from T. vivax across SA and WA support clonal propagation, and is consistent with the hypothesis that the SA populations examined here derived from common ancestors recently introduced from West Africa. The molecular markers defined here are valuable to assess the genetic diversity, to track the source and dispersion of outbreaks, and to explore the epidemiological and pathological significance of T. vivax genotypes.


Trypanosoma vivax/genetics , Trypanosomiasis, African/parasitology , Africa, Western/epidemiology , Animals , Genetic Variation , Genotype , Livestock , Microsatellite Repeats , Phylogeny , South America/epidemiology , Trypanosomiasis, African/epidemiology , Trypanosomiasis, African/genetics , Trypanosomiasis, African/mortality
14.
Exp Parasitol ; 135(2): 331-6, 2013 Oct.
Article En | MEDLINE | ID: mdl-23916765

Trypanosomosis has been associated with immunosuppression, anemia and oxidative damage while selenium possesses both immunostimulatory and antioxidative effects. This study was designed to assess the effect of dietary selenium supplementation on parasitemia, anemia, survival pattern and serum protein profiles of trypanosome-infected rats. Twenty five rats, divided into five groups (A-E) of 5 each, were treated as follows: 4, 8 and 16 ppm (ppm) of selenium in their feed, respectively throughout the experimental period and were infected with Trypanosoma brucei brucei on day 14 post supplementation, infected not supplemented and the negative control. Supplementation at 4 and 8 ppm increased the packed cell volume (PCV) and hemoglobin (Hb) concentration on day 7 of supplementation (PS) when compared with the unsupplemented groups. Following infection on day 14 PS, the PCV, Hb of 16 ppm and infected not supplemented groups were significantly (P < 0.05) lower than other groups on days 28 and 35 PS. Supplementation did not lead to significant (P > 0.05) changes on the total protein, albumin and globulin by day 14 PS. Infection, however, caused significant (P > 0.05) decrease in the total protein and albumin from day 28. The supplementation did not significantly (P > 0.05) increase the pre-patent period but caused a significant reduction in the parasitemia levels and increased survival intervals. Dietary selenium supplementation, from the results, may show promise in the management of African trypanosomosis as the supplementation was able to: reduce anemia and parasitemia and increase survival intervals of trypanosome infected rats.


Anemia/prevention & control , Blood Proteins/drug effects , Dietary Supplements , Sodium Selenite/administration & dosage , Trypanosoma brucei brucei , Trypanosomiasis, African/blood , Animals , Globulins/drug effects , Hematocrit , Hemoglobins/drug effects , Male , Parasitemia/prevention & control , Random Allocation , Rats , Serum Albumin/drug effects , Trypanosomiasis, African/complications , Trypanosomiasis, African/mortality
16.
Mol Microbiol ; 86(1): 51-64, 2012 Oct.
Article En | MEDLINE | ID: mdl-22857512

Pyridoxal-5'-phosphate (vitamin B(6) ) is an essential cofactor for many important enzymatic reactions such as transamination and decarboxylation. African trypanosomes are unable to synthesise vitamin B(6) de novo and rely on uptake of B(6) vitamers such as pyridoxal and pyridoxamine from their hosts, which are subsequently phosphorylated by pyridoxal kinase (PdxK). A conditional null mutant of PdxK was generated in Trypanosoma brucei bloodstream forms showing that this enzyme is essential for growth of the parasite in vitro and for infectivity in mice. Activity of recombinant T. brucei PdxK was comparable to previously published work having a specific activity of 327 ± 13 mU mg(-1) and a K(m)(app) with respect to pyridoxal of 29.6 ± 3.9 µM. A coupled assay was developed demonstrating that the enzyme has equivalent catalytic efficiency with pyridoxal, pyridoxamine and pyridoxine, and that ginkgotoxin is an effective pseudo substrate. A high resolution structure of PdxK in complex with ATP revealed important structural differences with the human enzyme. These findings suggest that pyridoxal kinase is an essential and druggable target that could lead to much needed alternative treatments for this devastating disease.


Pyridoxal Kinase/chemistry , Pyridoxal Kinase/genetics , Trypanosoma brucei brucei/enzymology , Amino Acid Sequence , Animals , Antiprotozoal Agents/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Deletion , Genes, Essential , Genes, Protozoan , Humans , Kinetics , Mice , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Conformation , Pyridoxal/metabolism , Pyridoxal Kinase/antagonists & inhibitors , Pyridoxamine/metabolism , Pyridoxine/analogs & derivatives , Pyridoxine/metabolism , Sequence Alignment , Survival Analysis , Trypanosoma brucei brucei/growth & development , Trypanosoma brucei brucei/pathogenicity , Trypanosomiasis, African/mortality , Trypanosomiasis, African/parasitology , Virulence Factors/antagonists & inhibitors , Virulence Factors/chemistry , Virulence Factors/genetics
17.
Adv Parasitol ; 79: 299-337, 2012.
Article En | MEDLINE | ID: mdl-22726645

Sleeping sickness describes two diseases, both fatal if left untreated: (i) Gambian sleeping sickness caused by Trypanosoma brucei gambiense, a chronic disease with average infection lasting around 3 years, and (ii) Rhodesian sleeping sickness caused by T. b. rhodesiense, an acute disease with death occurring within weeks of infection. Control of Gambian sleeping sickness is based on case detection and treatment involving serological screening, followed by diagnostic confirmation and staging. In stage I, patients can remain asymptomatic as trypanosomes multiply in tissues and body fluids; in stage II, trypanosomes cross the blood-brain barrier, enter the central nervous system and, if left untreated, death follows. Staging is crucial as it defines the treatment that is prescribed; for both forms of disease, stage II involves the use of the highly toxic drug melarsoprol or, in the case of Gambian sleeping sickness, the use of complex and very expensive drug regimes. Case detection of T. b. gambiense sleeping sickness is known to be inefficient but could be improved by the identification of parasites using molecular tools that are, as yet, rarely used in the field. Diagnostics are not such a problem in relation to T. b. rhodesiense sleeping sickness, but the high level of under-reporting of this disease suggests that current strategies, reliant on self-reporting, are inefficient. Sleeping sickness is one of the 'neglected tropical diseases' that attracts little attention from donors or policymakers. Proper quantification of the burden of sleeping sickness matters, as the primary reason for its 'neglect' is that the true impact of the disease is unknown, largely as a result of under-reporting. Certainly, elimination will not be achieved without vast improvements in field diagnostics for both forms of sleeping sickness especially if there is a hidden reservoir of 'chronic carriers'. Mass screening would be a desirable aim for Gambian sleeping sickness and could be handled on a national scale in the endemic countries - perhaps by piggybacking on programmes committed to other diseases. As well as improved diagnostics, the search for non-toxic drugs for stage II treatment should remain a research priority. There is good evidence that thorough active case finding is sufficient to control T. b. gambiense sleeping sickness, as there is no significant animal reservoir. Trypanosoma brucei rhodesiense sleeping sickness is a zoonosis and control involves interrupting the fly-animal-human cycle, so some form of tsetse control and chemotherapy of the animal reservoir must be involved. The restricted application of insecticide to cattle is the most promising, affordable and sustainable technique to have emerged for tsetse control. Animal health providers can aid disease control by treating cattle and, when allied with innovative methods of funding (e.g. public-private partnerships) not reliant on the public purse, this approach may prove more sustainable. Sleeping sickness incidence for the 36 endemic countries has shown a steady decline in recent years and we should take advantage of the apparent lull in incidence and aim for elimination. This is feasible in some sleeping sickness foci but must be planned and paid for increasingly by the endemic countries themselves. The control and elimination of T. b. gambiense sleeping sickness may be seen as a public good, as appropriate strategies depend on local health services for surveillance and treatment, but public-private funding mechanisms should not be excluded. It is timely to take up the tools available and invest in new tools - including novel financial instruments - to eliminate this disease from Africa.


Disease Eradication/methods , Trypanosoma brucei gambiense/pathogenicity , Trypanosoma brucei rhodesiense/pathogenicity , Trypanosomiasis, African/epidemiology , Trypanosomiasis, African/prevention & control , Africa/epidemiology , Animals , Cattle , Communicable Disease Control/methods , Humans , Incidence , Neglected Diseases/diagnosis , Neglected Diseases/epidemiology , Neglected Diseases/mortality , Neglected Diseases/prevention & control , Trypanosomiasis, African/diagnosis , Trypanosomiasis, African/mortality , Zoonoses/epidemiology , Zoonoses/parasitology , Zoonoses/transmission
18.
PLoS Negl Trop Dis ; 6(6): e1691, 2012.
Article En | MEDLINE | ID: mdl-22720107

The final outcome of infection by Trypanosoma brucei gambiense, the main agent of sleeping sickness, has always been considered as invariably fatal. While scarce and old reports have mentioned cases of self-cure in untreated patients, these studies suffered from the lack of accurate diagnostic tools available at that time. Here, using the most specific and sensitive tools available to date, we report on a long-term follow-up (15 years) of a cohort of 50 human African trypanosomiasis (HAT) patients from the Ivory Coast among whom 11 refused treatment after their initial diagnosis. In 10 out of 11 subjects who continued to refuse treatment despite repeated visits, parasite clearance was observed using both microscopy and polymerase chain reaction (PCR). Most of these subjects (7/10) also displayed decreasing serological responses, becoming progressively negative to trypanosome variable antigens (LiTat 1.3, 1.5 and 1.6). Hence, in addition to the "classic" lethal outcome of HAT, we show that alternative natural progressions of HAT may occur: progression to an apparently aparasitaemic and asymptomatic infection associated with strong long-lasting serological responses and progression to an apparently spontaneous resolution of infection (with negative results in parasitological tests and PCR) associated with a progressive drop in antibody titres as observed in treated cases. While this study does not precisely estimate the frequency of the alternative courses for this infection, it is noteworthy that in the field national control programs encounter a significant proportion of subjects displaying positive serologic test results but negative results in parasitological testing. These findings demonstrate that a number of these subjects display such infection courses. From our point of view, recognising that trypanotolerance exists in humans, as is now widely accepted for animals, is a major step forward for future research in the field of HAT.


Trypanosoma brucei gambiense/isolation & purification , Trypanosoma brucei gambiense/pathogenicity , Trypanosomiasis, African/mortality , Trypanosomiasis, African/parasitology , Animals , Antibodies, Protozoan/blood , Cohort Studies , Cote d'Ivoire , Female , Follow-Up Studies , Humans , Male , Microscopy/methods , Molecular Diagnostic Techniques/methods , Parasitology/methods , Survival Analysis , Treatment Refusal
20.
Infect Immun ; 77(9): 3948-57, 2009 Sep.
Article En | MEDLINE | ID: mdl-19596769

Trypanosoma congolense is a protozoan parasite that causes severe diseases in livestock. Three major quantative trait loci (QTL), Tir1, Tir2, and Tir3, control the survival time of mice after infection with T. congolense. Congenic mice carrying the C57BL/6 resistance alleles on the A/J background were developed for each of these loci. The congenic mice were used to physically map the regions containing the QTL gene(s) and to investigate the physiological effect of each locus. Clinical chemistry data for infected A/J, C57BL/6, and BALB/c mice were obtained for 15 analytes at five time points. Congenic mice were assessed for survival, parasitemia, and anemia as well as seven clinical-chemical analytes. The survival times were significantly increased in the Tir1 and Tir2 mice but not Tir3 congenic mice. The survival time of the parental inbred mice correlated negatively with parasitemia but positively with alanine aminotransferase activities in serum, suggesting that inflammatory reactions in the liver had a beneficial effect possibly associated with reduced parasitemia. However, there was no difference in parasitemia or liver enzyme activities of Tir1 and Tir2 congenic mice relative to their controls, showing that survival, parasitemia, and degree of liver damage are not associated with each other, despite the correlation in the parental lines. These data suggest that the congenic loci affect survival but do not affect control of parasite number. They may therefore act by limiting the pathological consequences of T. congolense infection.


Quantitative Trait Loci , Trypanosoma congolense , Trypanosomiasis, African/genetics , Alanine Transaminase/blood , Anemia/etiology , Animals , Electrolytes/blood , Haplotypes , Hemoglobins/analysis , Kidney/physiopathology , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Parasitemia/genetics , Trypanosomiasis, African/blood , Trypanosomiasis, African/mortality , Uric Acid/blood , alpha-Amylases/blood
...