Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Annu Rev Pathol ; 18: 19-45, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36055769

RESUMEN

African trypanosomes are bloodstream protozoan parasites that infect mammals including humans, where they cause sleeping sickness. Long-lasting infection is required to favor parasite transmission between hosts. Therefore, trypanosomes have developed strategies to continuously escape innate and adaptive responses of the immune system, while also preventing premature death of the host. The pathology linked to infection mainly results from inflammation and includes anemia and brain dysfunction in addition to loss of specificity and memory of the antibody response. The serum of humans contains an efficient trypanolytic factor, the membrane pore-forming protein apolipoprotein L1 (APOL1). In the two human-infective trypanosomes, specific parasite resistance factors inhibit APOL1 activity. In turn, many African individuals express APOL1 variants that counteract these resistance factors, enabling them to avoid sleeping sickness. However, these variants are associated with chronic kidney disease, particularly in the context of virus-induced inflammation such as coronavirus disease 2019. Vaccination perspectives are discussed.


Asunto(s)
COVID-19 , Tripanosomiasis Africana , Humanos , Apolipoproteína L1/genética , Inflamación , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/genética , Tripanosomiasis Africana/parasitología
2.
PLoS Negl Trop Dis ; 12(11): e0006905, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30408045

RESUMEN

This paper presents the development of an agent-based model (ABM) to investigate Trypanosoma brucei rhodesiense human African trypanosomiasis (rHAT) disease transmission. The ABM model, fitted at a fine spatial scale, was used to explore the impact of a growing host population on the spread of disease along a 75 km transect in the Luangwa Valley, Zambia. The model was used to gain a greater understanding of how increases in human and domestic animal population could impact the contact network between vector and host, the subsequent transmission patterns, and disease incidence outcomes in the region. Modelled incidence rates showed increases in rHAT transmission in both humans and cattle. The primary demographic attribution of infection switched dramatically from young children of both sexes attending school, to adult women performing activities with shorter but more frequent trips, such as water and firewood collection, with men more protected due to the presence of cattle in their routines. The interpretation of model output provides a plausible insight into both population development and disease transmission in the near future in the region and such techniques could aid well-targeted mitigation strategies in the future.


Asunto(s)
Enfermedades de los Bovinos/transmisión , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/transmisión , Adolescente , Adulto , Anciano , Animales , Bovinos , Enfermedades de los Bovinos/epidemiología , Enfermedades de los Bovinos/parasitología , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Modelos Biológicos , Crecimiento Demográfico , Tripanosomiasis Africana/epidemiología , Tripanosomiasis Africana/etnología , Tripanosomiasis Africana/parasitología , Adulto Joven , Zambia/epidemiología , Zambia/etnología
4.
PLoS One ; 11(10): e0161291, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27706167

RESUMEN

BACKGROUND: Identifying hosts of blood-feeding insect vectors is crucial in understanding their role in disease transmission. Rhodesian human African trypanosomiasis (rHAT), also known as acute sleeping sickness is caused by Trypanosoma brucei rhodesiense and transmitted by tsetse flies. The disease is commonly associated with wilderness areas of east and southern Africa. Such areas hold a diverse range of species which form communities of hosts for disease maintenance. The relative importance of different wildlife hosts remains unclear. This study quantified tsetse feeding preferences in a wilderness area of great host species richness, Serengeti National Park, Tanzania, assessing tsetse feeding and host density contemporaneously. METHODS: Glossina swynnertoni and G. pallidipes were collected from six study sites. Bloodmeal sources were identified through matching Cytochrome B sequences amplified from bloodmeals from recently fed flies to published sequences. Densities of large mammal species in each site were quantified, and feeding indices calculated to assess the relative selection or avoidance of each host species by tsetse. RESULTS: The host species most commonly identified in G. swynnertoni bloodmeals, warthog (94/220), buffalo (48/220) and giraffe (46/220), were found at relatively low densities (3-11/km2) and fed on up to 15 times more frequently than expected by their relative density. Wildebeest, zebra, impala and Thomson's gazelle, found at the highest densities, were never identified in bloodmeals. Commonly identified hosts for G. pallidipes were buffalo (26/46), giraffe (9/46) and elephant (5/46). CONCLUSIONS: This study is the first to quantify tsetse host range by molecular analysis of tsetse diet with simultaneous assessment of host density in a wilderness area. Although G. swynnertoni and G. pallidipes can feed on a range of species, they are highly selective. Many host species are rarely fed on, despite being present in areas where tsetse are abundant. These feeding patterns, along with the ability of key host species to maintain and transmit T. b. rhodesiense, drive the epidemiology of rHAT in wilderness areas.


Asunto(s)
Interacciones Huésped-Parásitos/fisiología , Insectos Vectores/parasitología , Moscas Tse-Tse/parasitología , Animales , Citocromos b/química , Citocromos b/genética , Citocromos b/metabolismo , Bases de Datos Genéticas , Conducta Alimentaria/fisiología , Femenino , Humanos , Masculino , Mamíferos/genética , Mamíferos/parasitología , Parques Recreativos , Tanzanía , Trypanosoma brucei rhodesiense/aislamiento & purificación , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/parasitología , Moscas Tse-Tse/genética , Moscas Tse-Tse/metabolismo
5.
Arch Pharm Res ; 39(10): 1391-1403, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27585596

RESUMEN

3-Azabicyclo[3.2.2]nonanes are already reported as antiprotozoal agents. Structural variations were performed by attachment of several basic side chains, being part of drugs in use, to the ring nitrogen. The structures of the new compounds were established using one and two dimensional NMR measurements. All compounds were investigated for their antiplasmodial and antitrypanosomal activities against Plasmodium falciparum K 1 (multiresistant) and Trypanosoma brucei rhodesiense. Their cytotoxicity was assessed against L6 cells. The results are compared to the activities of formerly synthesized compounds. Structure-activity relationships are discussed.


Asunto(s)
Alcanos/síntesis química , Alcanos/farmacología , Antiprotozoarios/síntesis química , Antiprotozoarios/farmacología , Compuestos de Azabiciclo/síntesis química , Compuestos de Azabiciclo/farmacología , Animales , Humanos , Ratones , Pruebas de Sensibilidad Parasitaria/métodos , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/fisiología , Relación Estructura-Actividad , Trypanosoma brucei rhodesiense/efectos de los fármacos , Trypanosoma brucei rhodesiense/fisiología
6.
Trends Parasitol ; 32(8): 608-621, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27262917

RESUMEN

Many wilderness areas of East and Southern Africa are foci for Rhodesian sleeping sickness, a fatal zoonotic disease caused by trypanosomes transmitted by tsetse flies. Although transmission in these foci is traditionally driven by wildlife reservoirs, rising human and livestock populations may increase the role of livestock in transmission cycles. Deciphering transmission dynamics at wildlife and livestock interface areas is key to developing appropriate control. Data are lacking for key parameters, including host distributions, tsetse density, and mortality rates, and the relative roles of livestock and wildlife as hosts in fragmented habitats, limiting the development of meaningful models to assist in the assessment and implementation of control strategies.


Asunto(s)
Animales Salvajes , Ganado , Tripanosomiasis Africana/transmisión , Distribución Animal , Animales , Ecosistema , Densidad de Población , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/prevención & control , Moscas Tse-Tse/parasitología
7.
Curr Opin Microbiol ; 32: 26-30, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27131101

RESUMEN

African trypanosomes, which divide their life cycle between mammals and tsetse flies, are confronted with environments that differ widely in temperature, nutrient availability and host responses to infection. In particular, since trypanosomes cannot predict when they will be transmitted between hosts, it is vital for them to be able to sense and adapt to their milieu. Thanks to technical advances, significant progress has been made in understanding how the parasites perceive external stimuli and react to them. There is also a growing awareness that trypanosomes use a variety of mechanisms to exchange information with each other, thereby enhancing their chances of survival.


Asunto(s)
Ambiente , Interacciones Huésped-Patógeno/fisiología , Estadios del Ciclo de Vida/fisiología , Mamíferos/parasitología , Trypanosoma brucei gambiense/fisiología , Trypanosoma brucei rhodesiense/fisiología , Moscas Tse-Tse/parasitología , Animales , Insectos Vectores/parasitología , Percepción de Quorum/fisiología , Trypanosoma brucei brucei/patogenicidad , Trypanosoma brucei gambiense/patogenicidad , Trypanosoma brucei rhodesiense/patogenicidad , Tripanosomiasis Africana/parasitología
8.
mBio ; 7(2): e02198-15, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27073096

RESUMEN

UNLABELLED: African trypanosomes, except Trypanosoma brucei gambiense and Trypanosoma brucei rhodesiense, which cause human African trypanosomiasis, are lysed by the human serum protein apolipoprotein L1 (ApoL1). These two subspecies can resist human ApoL1 because they express the serum resistance proteins T. b. gambiense glycoprotein (TgsGP) and serum resistance-associated protein (SRA), respectively. Whereas in T. b. rhodesiense, SRA is necessary and sufficient to inhibit ApoL1, in T. b. gambiense, TgsGP cannot protect against high ApoL1 uptake, so different additional mechanisms contribute to limit this uptake. Here we report a complex interplay between trypanosomes and an ApoL1 variant, revealing important insights into innate human immunity against these parasites. Using whole-genome sequencing, we characterized an atypical T. b. gambiense infection in a patient in Ghana. We show that the infecting trypanosome has diverged from the classical T. b. gambiense strains and lacks the TgsGP defense mechanism against human serum. By sequencing the ApoL1 gene of the patient and subsequent in vitro mutagenesis experiments, we demonstrate that a homozygous missense substitution (N264K) in the membrane-addressing domain of this ApoL1 variant knocks down the trypanolytic activity, allowing the trypanosome to avoid ApoL1-mediated immunity. IMPORTANCE: Most African trypanosomes are lysed by the ApoL1 protein in human serum. Only the subspecies Trypanosoma b. gambiense and T. b. rhodesiense can resist lysis by ApoL1 because they express specific serum resistance proteins. We here report a complex interplay between trypanosomes and an ApoL1 variant characterized by a homozygous missense substitution (N264K) in the domain that we hypothesize interacts with the endolysosomal membranes of trypanosomes. The N264K substitution knocks down the lytic activity of ApoL1 against T. b. gambiense strains lacking the TgsGP defense mechanism and against T. b. rhodesiense if N264K is accompanied by additional substitutions in the SRA-interacting domain. Our data suggest that populations with high frequencies of the homozygous N264K ApoL1 variant may be at increased risk of contracting human African trypanosomiasis.


Asunto(s)
Apolipoproteínas/genética , Susceptibilidad a Enfermedades , Variación Genética , Lipoproteínas HDL/genética , Trypanosoma brucei gambiense/fisiología , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/genética , Apolipoproteína L1 , Apolipoproteínas/inmunología , Humanos , Inmunidad Innata , Lipoproteínas HDL/inmunología , Mutación Missense , Polimorfismo de Nucleótido Simple , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Tripanosomiasis Africana/inmunología , Tripanosomiasis Africana/parasitología
9.
PLoS One ; 11(2): e0147660, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26910229

RESUMEN

BACKGROUND: Trypanosoma brucei is a eukaryotic pathogen which causes African trypanosomiasis. It is notable for its variant surface glycoprotein (VSG) coat, which undergoes antigenic variation enabled by a large suite of VSG pseudogenes, allowing for persistent evasion of host adaptive immunity. While Trypanosoma brucei rhodesiense (Tbr) and T. b gambiense (Tbg) are human infective, related T. b. brucei (Tbb) is cleared by human sera. A single gene, the Serum Resistance Associated (SRA) gene, confers Tbr its human infectivity phenotype. Potential genetic recombination of this gene between Tbr and non-human infective Tbb strains has significant epidemiological consequences for Human African Trypanosomiasis outbreaks. RESULTS: Using long and short read whole genome sequencing, we generated a hybrid de novo assembly of a Tbr strain, producing 4,210 scaffolds totaling approximately 38.8 megabases, which comprise a significant proportion of the Tbr genome, and thus represents a valuable tool for a comparative genomics analyses among human and non-human infective T. brucei and future complete genome assembly. We detected 5,970 putative genes, of which two, an alcohol oxidoreductase and a pentatricopeptide repeat-containing protein, were members of gene families common to all T. brucei subspecies, but variants specific to the Tbr strain sequenced in this study. Our findings confirmed the extremely high level of genomic similarity between the two parasite subspecies found in other studies. CONCLUSIONS: We confirm at the whole genome level high similarity between the two Tbb and Tbr strains studied. The discovery of extremely minor genomic differentiation between Tbb and Tbr suggests that the transference of the SRA gene via genetic recombination could potentially result in novel human infective strains, thus all genetic backgrounds of T. brucei should be considered potentially human infective in regions where Tbr is prevalent.


Asunto(s)
Genómica , Trypanosoma brucei brucei/genética , Trypanosoma brucei rhodesiense/genética , Evolución Molecular , Transferencia de Gen Horizontal , Humanos , Glicoproteínas de Membrana/genética , Proteínas Protozoarias/genética , Análisis de Secuencia , Trypanosoma brucei brucei/fisiología , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/epidemiología
10.
Mol Microbiol ; 97(3): 397-407, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25899052

RESUMEN

Human apolipoprotein L1 (APOL1) kills African trypanosomes except Trypanosoma rhodesiense and Trypanosoma gambiense, the parasites causing sleeping sickness. APOL1 uptake into trypanosomes is favoured by its association with the haptoglobin-related protein-haemoglobin complex, which binds to the parasite surface receptor for haptoglobin-haemoglobin. As haptoglobin-haemoglobin can saturate the receptor, APOL1 uptake is increased in haptoglobin-poor (hypohaptoglobinaemic) serum (HyHS). While T. rhodesiense resists APOL1 by RNA polymerase I (pol-I)-mediated expression of the serum resistance-associated (SRA) protein, T. gambiense resists by pol-II-mediated expression of the T. gambiense-specific glycoprotein (TgsGP). Moreover, in T. gambiense resistance to HyHS is linked to haptoglobin-haemoglobin receptor inactivation by mutation. We report that unlike T. gambiense, T. rhodesiense possesses a functional haptoglobin-haemoglobin receptor, and that like T. gambiense experimentally provided with active receptor, this parasite is killed in HyHS because of receptor-mediated APOL1 uptake. However, T. rhodesiense could adapt to low haptoglobin by increasing transcription of SRA. When assayed in Trypanosoma brucei, resistance to HyHS occurred with pol-I-, but not with pol-II-mediated SRA expression. Similarly, T. gambiense provided with active receptor acquired resistance to HyHS only when TgsGP was moved to a pol-I locus. Thus, transcription by pol-I favours adaptive gene regulation, explaining the presence of SRA in a pol-I locus.


Asunto(s)
Apolipoproteínas/toxicidad , Regulación de la Expresión Génica , Lipoproteínas HDL/toxicidad , ARN Polimerasa I/metabolismo , Transcripción Genética , Trypanosoma brucei rhodesiense/efectos de los fármacos , Trypanosoma brucei rhodesiense/fisiología , Adaptación Fisiológica , Apolipoproteína L1 , Haptoglobinas/análisis , Humanos , Glicoproteínas de Membrana/biosíntesis , Receptores de Superficie Celular/metabolismo , Suero/química , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/crecimiento & desarrollo , Trypanosoma brucei gambiense/efectos de los fármacos , Trypanosoma brucei gambiense/genética , Trypanosoma brucei gambiense/crecimiento & desarrollo , Trypanosoma brucei rhodesiense/genética , Trypanosoma brucei rhodesiense/crecimiento & desarrollo
11.
Arch Pharm Res ; 38(8): 1455-67, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25433423

RESUMEN

Some antimalarial agents in use typically bear basic side chains as ligands. Such ligands were attached to the amino substituent of a bridgehead atom of already antiprotozoal active 3-azabicyclo[3.2.2]nonanes. Structure verification was done by NMR measurements. The new compounds were tested for their antiplasmodial and antitrypanosomal activities against Plasmodium falciparum K 1 (multiresistant) and Trypanosoma brucei rhodesiense as well as for their cytotoxicity against L6 cells. Their activities are compared to those of already prepared compounds and structure-activity relationships are discussed.


Asunto(s)
Alcanos/síntesis química , Antiprotozoarios/síntesis química , Compuestos de Azabiciclo/síntesis química , Plasmodium falciparum/efectos de los fármacos , Trypanosoma brucei rhodesiense/efectos de los fármacos , Alcanos/farmacología , Antiprotozoarios/farmacología , Compuestos de Azabiciclo/farmacología , Humanos , Plasmodium falciparum/fisiología , Trypanosoma brucei rhodesiense/fisiología
12.
J Insect Sci ; 142014.
Artículo en Inglés | MEDLINE | ID: mdl-25527583

RESUMEN

The establishment of infection with three Trypanosoma spp (Gruby) (Kinetoplastida: Trypanosomatidae), specifically Trypanosoma brucei brucei (Plimmer and Bradford), T. b. rhodesiense (Stephen and Fatham) and T. congolense (Broden) was evaluated in Glossina pallidipes (Austen) (Diptera: Glossinidae) that either harbored or were uninfected by the endosymbiont Sodalis glossinidius (Dale and Maudlin) (Enterobacteriales: Enterobacteriaceae). Temporal variation of co-infection with T. b. rhodesiense and S. glossinidius was also assessed. The results show that both S. glossinidius infection (χ(2)= 1.134, df = 2, P = 0.567) and trypanosome infection rate (χ(2)= 1.85, df = 2, P = 0.397) were comparable across the three infection groups. A significant association was observed between the presence of S. glossinidius and concurrent trypanosome infection with T. b. rhodesiense (P = 0.0009) and T. congolense (P = 0.0074) but not with T. b. brucei (P = 0.5491). The time-series experiment revealed a slight decrease in the incidence of S. glossinidius infection with increasing fly age, which may infer a fitness cost associated with Sodalis infection. The present findings contribute to research on the feasibility of S. glossinidius-based paratransgenic approaches in tsetse and trypanosomiasis control, in particular relating to G. pallidipes control.


Asunto(s)
Enterobacteriaceae/fisiología , Insectos Vectores/microbiología , Insectos Vectores/fisiología , Trypanosoma/fisiología , Moscas Tse-Tse/microbiología , Moscas Tse-Tse/parasitología , Animales , Masculino , Especificidad de la Especie , Simbiosis , Trypanosoma brucei brucei/fisiología , Trypanosoma brucei rhodesiense/fisiología , Trypanosoma congolense/fisiología
13.
Trends Parasitol ; 29(8): 394-406, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23845311

RESUMEN

Uganda has both forms of human African trypanosomiasis (HAT): the chronic gambiense disease in the northwest and the acute rhodesiense disease in the south. The recent spread of rhodesiense into central Uganda has raised concerns given the different control strategies the two diseases require. We present knowledge on the population genetics of the major vector species Glossina fuscipes fuscipes in Uganda with a focus on population structure, measures of gene flow between populations, and the occurrence of polyandry. The microbiome composition and diversity is discussed, focusing on their potential role on trypanosome infection outcomes. We discuss the implications of these findings for large-scale tsetse control programs, including suppression or eradication, being undertaken in Uganda, and potential future genetic applications.


Asunto(s)
Insectos Vectores/fisiología , Control Biológico de Vectores/métodos , Trypanosoma brucei gambiense/fisiología , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/transmisión , Moscas Tse-Tse/fisiología , Animales , Biodiversidad , ADN Mitocondrial/genética , Femenino , Flujo Génico , Genética de Población , Interacciones Huésped-Patógeno , Humanos , Insectos Vectores/genética , Insectos Vectores/parasitología , Masculino , Microbiota , Repeticiones de Microsatélite/genética , Simbiosis , Tripanosomiasis Africana/epidemiología , Tripanosomiasis Africana/parasitología , Moscas Tse-Tse/genética , Moscas Tse-Tse/parasitología , Uganda/epidemiología
14.
Rev Neurol (Paris) ; 168(3): 230-8, 2012 Mar.
Artículo en Francés | MEDLINE | ID: mdl-22398218

RESUMEN

Sleeping sickness or human African trypanosomiasis is a parasitic disease transmitted by tsetse flies and therefore confined to its habitat, the central part of the African continent. Two disease forms are linked to two different parasites: T. b. gambiense and T. b. rhodesiense. Actual epidemiological data and precise and dynamic mapping of foci are in favor of a real decrease of the disease. Not all areas are under control and resurgence can still not be avoided from the remote areas where the disease is endemic. However, recent advances in knowledge in parasite genetics are giving hope of control. In 2009, for the first time since 50 years, less than 10,000 cases were declared to the World Health Organization. Clinical trials allowed revising some clinical concepts and linking them with parasite genetics: both disease forms can show variations from asymptomatic, chronic to acute and are linked to genetic differences in the host or the parasite. Parasitological diagnosis may be facilitated by the introduction of individual rapid tests and PCR-based field tests. Knowledge in mechanisms of brain invasion and screenings of inflammatory molecules allow new marker combinations for staging but they do not avoid lumbar puncture. Therapeutic options remain limited but there is hope to develop a new drug orally available in a near future.


Asunto(s)
Tripanosomiasis Africana/epidemiología , África/epidemiología , Animales , Epidemias , Humanos , Reacción en Cadena de la Polimerasa , Trypanosoma brucei gambiense/genética , Trypanosoma brucei gambiense/fisiología , Trypanosoma brucei rhodesiense/genética , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/parasitología , Tripanosomiasis Africana/patología , Tripanosomiasis Africana/terapia
15.
Microbiology (Reading) ; 157(Pt 10): 2933-2941, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21737496

RESUMEN

The transmigration of African trypanosomes across the human blood-brain barrier (BBB) is the critical step during the course of human African trypanosomiasis. The parasites Trypanosoma brucei gambiense and T. b. rhodesiense are transmitted to humans during the bite of tsetse flies. Trypanosomes multiply within the bloodstream and finally invade the central nervous system (CNS), which leads to the death of untreated patients. This project focused on the mechanisms of trypanosomal traversal across the BBB. In order to establish a suitable in vitro BBB model for parasite transmigration, different human cell lines were used, including ECV304, HBMEC and HUVEC, as well as C6 rat astrocytes. Validation of the BBB models with Escherichia coli HB101 and E. coli K1 revealed that a combination of ECV304 cells seeded on Matrigel as a semi-synthetic basement membrane and C6 astrocytes resulted in an optimal BBB model system. The BBB model showed selective permeability for the pathogenic E. coli K1 strain, and African trypanosomes were able to traverse the optimized ECV304-C6 BBB efficiently. Furthermore, coincubation indicated that paracellular macrophage transmigration does not facilitate trypanosomal BBB traversal. An inverse assembly of the BBB model demonstrated that trypanosomes were also able to transmigrate the optimized ECV304-C6 BBB backwards, indicating the relevance of the CNS as a possible reservoir of a relapsing parasitaemia.


Asunto(s)
Barrera Hematoencefálica/parasitología , Modelos Biológicos , Trypanosoma brucei gambiense/fisiología , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/parasitología , Animales , Línea Celular , Humanos , Ratones , Ratas
16.
Eukaryot Cell ; 10(8): 1023-33, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21705681

RESUMEN

Trypanosoma brucei rhodesiense is the causative agent of human African sleeping sickness. While the closely related subspecies T. brucei brucei is highly susceptible to lysis by a subclass of human high-density lipoproteins (HDL) called trypanosome lytic factor (TLF), T. brucei rhodesiense is resistant and therefore able to establish acute and fatal infections in humans. This resistance is due to expression of the serum resistance-associated (SRA) gene, a member of the variant surface glycoprotein (VSG) gene family. Although much has been done to establish the role of SRA in human serum resistance, the specific molecular mechanism of SRA-mediated resistance remains a mystery. Thus, we report the trafficking and steady-state localization of SRA in order to provide more insight into the mechanism of SRA-mediated resistance. We show that SRA traffics to the flagellar pocket of bloodstream-form T. brucei organisms, where it localizes transiently before being endocytosed to its steady-state localization in endosomes, and we demonstrate that the critical point of colocalization between SRA and TLF occurs intracellularly.


Asunto(s)
Endosomas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Protozoarias/metabolismo , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/parasitología , Células Cultivadas , Flagelos/metabolismo , Humanos , Evasión Inmune , Lipoproteínas HDL/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Trypanosoma brucei rhodesiense/metabolismo , Tripanosomiasis Africana/inmunología
17.
Int J Infect Dis ; 15(8): e517-24, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21683638

RESUMEN

Human African trypanosomiasis (HAT) is caused by sub-species of the parasitic protozoan Trypanosoma brucei and is transmitted by tsetse flies, both of which are endemic only to sub-Saharan Africa. Several cases have been reported in non-endemic areas, such as North America and Europe, due to travelers, ex-patriots or military personnel returning from abroad or due to immigrants from endemic areas. In this paper, non-endemic cases reported over the past 20 years are reviewed; a total of 68 cases are reported, 19 cases of Trypanosoma brucei gambiense HAT and 49 cases of Trypanosoma brucei rhodesiense HAT. Patients ranged in age from 19 months to 72 years and all but two patients survived. Physicians in non-endemic areas should be aware of the signs and symptoms of this disease, as well as methods of diagnosis and treatment, especially as travel to HAT endemic areas increases. We recommend extension of the current surveillance systems such as TropNetEurop and maintaining and promotion of existing reference centers of diagnostics and expertise. Important contact information is also included, should physicians require assistance in diagnosing or treating HAT.


Asunto(s)
Vigilancia de la Población , Tripanosomiasis Africana/epidemiología , Adolescente , Adulto , Anciano , Animales , Niño , Preescolar , Europa (Continente)/epidemiología , Femenino , Humanos , Lactante , Insectos Vectores/parasitología , Masculino , Persona de Mediana Edad , América del Norte/epidemiología , Viaje , Trypanosoma brucei brucei/fisiología , Trypanosoma brucei gambiense/fisiología , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/parasitología , Tripanosomiasis Africana/terapia , Moscas Tse-Tse/parasitología , Adulto Joven
18.
Trends Parasitol ; 26(9): 457-64, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20646962

RESUMEN

The Trypanosoma brucei subspecies T. brucei brucei is non-human infective due to susceptibility to lysis by trypanolytic factor (TLF) in human serum. Reviewed here are the advances which have revealed apolipoprotein L1 (ApoL1), found in high density lipoprotein, as the lysis-inducing component of TLF, the means of uptake via haptoglobin-related protein receptor and the mechanism of resistance in T. b. rhodesiense via its serum resistance-associated (SRA) protein. The first practical steps to application of these discoveries are now in progress; transgenic animals expressing either baboon or minimally truncated human ApoL1 show resistance to both T. b. brucei and T. b. rhodesiense. This has major implications for treatment and prevention of human and animal African trypanosomiasis.


Asunto(s)
Apolipoproteínas/metabolismo , Interacciones Huésped-Parásitos , Lipoproteínas HDL/metabolismo , Trypanosoma brucei brucei/fisiología , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/inmunología , Animales , Antígenos de Neoplasias/metabolismo , Apolipoproteína L1 , Haptoglobinas/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Papio , Proteínas Protozoarias/metabolismo , Tripanosomiasis Africana/parasitología
19.
PLoS Pathog ; 5(12): e1000685, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19997494

RESUMEN

Apolipoprotein L-I (apoL1) is a human-specific serum protein that kills Trypanosoma brucei through ionic pore formation in endosomal membranes of the parasite. The T. brucei subspecies rhodesiense and gambiense resist this lytic activity and can infect humans, causing sleeping sickness. In the case of T. b. rhodesiense, resistance to lysis involves interaction of the Serum Resistance-Associated (SRA) protein with the C-terminal helix of apoL1. We undertook a mutational and deletional analysis of the C-terminal helix of apoL1 to investigate the linkage between interaction with SRA and lytic potential for different T. brucei subspecies. We confirm that the C-terminal helix is the SRA-interacting domain. Although in E. coli this domain was dispensable for ionic pore-forming activity, its interaction with SRA resulted in inhibition of this activity. Different mutations affecting the C-terminal helix reduced the interaction of apoL1 with SRA. However, mutants in the L370-L392 leucine zipper also lost in vitro trypanolytic activity. Truncating and/or mutating the C-terminal sequence of human apoL1 like that of apoL1-like sequences of Papio anubis resulted in both loss of interaction with SRA and acquired ability to efficiently kill human serum-resistant T. b. rhodesiense parasites, in vitro as well as in transgenic mice. These findings demonstrate that SRA interaction with the C-terminal helix of apoL1 inhibits its pore-forming activity and determines resistance of T. b. rhodesiense to human serum. In addition, they provide a possible explanation for the ability of Papio serum to kill T. b. rhodesiense, and offer a perspective to generate transgenic cattle resistant to both T. b. brucei and T. b. rhodesiense.


Asunto(s)
Apolipoproteínas/fisiología , Supervivencia Celular/efectos de los fármacos , Lipoproteínas HDL/fisiología , Glicoproteínas de Membrana/metabolismo , Proteínas Protozoarias/metabolismo , Trypanosoma brucei brucei/fisiología , Trypanosoma brucei rhodesiense/fisiología , Secuencia de Aminoácidos , Animales , Apolipoproteína L1 , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Apolipoproteínas/farmacología , Análisis Mutacional de ADN , Humanos , Leucina Zippers/genética , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , Lipoproteínas HDL/farmacología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Papio anubis , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteínas Citotóxicas Formadoras de Poros/farmacología , Unión Proteica , Alineación de Secuencia , Termodinámica , Tripanocidas/metabolismo , Tripanocidas/farmacología , Trypanosoma brucei brucei/metabolismo , Trypanosoma brucei rhodesiense/metabolismo
20.
Annu Rev Microbiol ; 63: 335-62, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19575562

RESUMEN

African trypanosomes are devastating human and animal pathogens. Trypanosoma brucei rhodesiense and T. b. gambiense subspecies cause the fatal human disease known as African sleeping sickness. It is estimated that several hundred thousand new infections occur annually and the disease is fatal if untreated. T. brucei is transmitted by the tsetse fly and alternates between bloodstream-form and insect-form life cycle stages that are adapted to survive in the mammalian host and the insect vector, respectively. The importance of the flagellum for parasite motility and attachment to the tsetse fly salivary gland epithelium has been appreciated for many years. Recent studies have revealed both conserved and novel features of T. brucei flagellum structure and composition, as well as surprising new functions that are outlined here. These discoveries are important from the standpoint of understanding trypanosome biology and identifying novel drug targets, as well as for advancing our understanding of fundamental aspects of eukaryotic flagellum structure and function.


Asunto(s)
Flagelos/fisiología , Locomoción , Trypanosoma brucei gambiense/fisiología , Trypanosoma brucei rhodesiense/fisiología , Animales , Flagelos/química , Flagelos/ultraestructura , Humanos , Moscas Tse-Tse
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...