Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Nat Microbiol ; 6(3): 392-400, 2021 03.
Article in English | MEDLINE | ID: mdl-33462435

ABSTRACT

Suramin has been a primary early-stage treatment for African trypanosomiasis for nearly 100 yr. Recent studies revealed that trypanosome strains that express the variant surface glycoprotein (VSG) VSGsur possess heightened resistance to suramin. Here, we show that VSGsur binds tightly to suramin but other VSGs do not. By solving high-resolution crystal structures of VSGsur and VSG13, we also demonstrate that these VSGs define a structurally divergent subgroup of the coat proteins. The co-crystal structure of VSGsur with suramin reveals that the chemically symmetric drug binds within a large cavity in the VSG homodimer asymmetrically, primarily through contacts of its central benzene rings. Structure-based, loss-of-contact mutations in VSGsur significantly decrease the affinity to suramin and lead to a loss of the resistance phenotype. Altogether, these data show that the resistance phenotype is dependent on the binding of suramin to VSGsur, establishing that the VSG proteins can possess functionality beyond their role in antigenic variation.


Subject(s)
Drug Resistance/immunology , Suramin/metabolism , Trypanosoma brucei rhodesiense/immunology , Variant Surface Glycoproteins, Trypanosoma/chemistry , Variant Surface Glycoproteins, Trypanosoma/metabolism , Antigenic Variation/drug effects , Antigenic Variation/immunology , Binding Sites , Crystallography, X-Ray , Drug Resistance/genetics , Endocytosis/genetics , Immune Evasion , Mutation , Protein Binding , Protein Conformation , Suramin/toxicity , Trypanocidal Agents/metabolism , Trypanocidal Agents/toxicity , Trypanosoma brucei rhodesiense/chemistry , Trypanosoma brucei rhodesiense/drug effects , Trypanosoma brucei rhodesiense/metabolism , Trypanosomiasis, African/parasitology , Variant Surface Glycoproteins, Trypanosoma/genetics
2.
J Med Chem ; 62(23): 10617-10629, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31714776

ABSTRACT

This paper describes an optimization strategy of the highly active vinyl ketone 3 which was recognized as a strong inhibitor of rhodesain of Trypanosoma brucei rhodesiense, endowed with a ksecond value of 67 × 106 M-1 min-1 coupled with a high binding affinity (Ki = 38 pM). We now report a new structure-activity relationship study based on structural variations on the P3, P2, and P1' sites which led us to identify two potent lead compounds, i.e., vinyl ketones 4h and 4k. Vinyl ketone 4h showed an impressive potency toward rhodesain (ksecond = 8811 × 105) coupled to a good antiparasitic activity (EC50 = 3.6 µM), while vinyl ketone 4k proved to possess the highest binding affinity toward the trypanosomal protease (Ki = 0.6 pM) and a submicromolar antiparasitic activity (EC50 = 0.67 µM), thus representing new lead compounds in the drug discovery process for the treatment of Human African Trypanosomiasis.


Subject(s)
Cysteine Endopeptidases/metabolism , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei rhodesiense/drug effects , Trypanosomiasis, African/drug therapy , Humans , Molecular Structure , Protein Conformation , Structure-Activity Relationship , Sulfones/chemistry , Sulfones/pharmacology , Trypanosoma brucei rhodesiense/metabolism
3.
ChemMedChem ; 13(20): 2150-2158, 2018 10 22.
Article in English | MEDLINE | ID: mdl-30152195

ABSTRACT

Trypanosoma brucei is the agent of human African trypanosomiasis (HAT), a neglected disease that threatens the lives of 65 million people in sub-Saharan Africa every year. Unfortunately, available therapies are unsatisfactory, due primarily to safety issues and development of drug resistance. Over the last decades significant effort has been made in the discovery of new potential anti-HAT agents, with help from the World Health Organization (WHO) and private-public partnerships such as the Drugs for Neglected Diseases Initiative (DNDi). Whereas antifolates have been a valuable source of drugs against bacterial infections and malaria, compounds effective against T. brucei have not yet been identified. Considering the relatively simple folate metabolic pathway in T. brucei, along with results obtained in this research field so far, we believe that further investigations might lead to effective chemotherapeutic agents. Herein we present a selection of the more promising results obtained so far in this field, underlining the opportunities that could lead to successful therapeutic approaches in the future.


Subject(s)
Folic Acid Antagonists/therapeutic use , Folic Acid/metabolism , Trypanocidal Agents/therapeutic use , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/drug therapy , Animals , Folic Acid Antagonists/pharmacology , Humans , Neglected Diseases/drug therapy , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/metabolism , Trypanosoma brucei rhodesiense/drug effects , Trypanosoma brucei rhodesiense/metabolism
4.
PLoS Negl Trop Dis ; 12(2): e0006280, 2018 02.
Article in English | MEDLINE | ID: mdl-29474390

ABSTRACT

All of our current knowledge of African trypanosome metabolism is based on results from trypanosomes grown in culture or in rodents. Drugs against sleeping sickness must however treat trypanosomes in humans. We here compare the transcriptomes of Trypanosoma brucei rhodesiense from the blood and cerebrospinal fluid of human patients with those of trypanosomes from culture and rodents. The data were aligned and analysed using new user-friendly applications designed for Kinetoplastid RNA-Seq data. The transcriptomes of trypanosomes from human blood and cerebrospinal fluid did not predict major metabolic differences that might affect drug susceptibility. Usefully, there were relatively few differences between the transcriptomes of trypanosomes from patients and those of similar trypanosomes grown in rats. Transcriptomes of monomorphic laboratory-adapted parasites grown in in vitro culture closely resembled those of the human parasites, but some differences were seen. In poly(A)-selected mRNA transcriptomes, mRNAs encoding some protein kinases and RNA-binding proteins were under-represented relative to mRNA that had not been poly(A) selected; further investigation revealed that the selection tends to result in loss of longer mRNAs.


Subject(s)
Gene Expression Profiling , RNA, Protozoan/isolation & purification , Transcriptome , Trypanosoma brucei rhodesiense/genetics , Trypanosoma brucei rhodesiense/isolation & purification , Trypanosomiasis, African/parasitology , Animals , Bacteriological Techniques/methods , DNA, Kinetoplast/genetics , Humans , Protein Kinases/genetics , Protozoan Proteins/genetics , RNA, Messenger/genetics , RNA, Protozoan/genetics , RNA-Binding Proteins/genetics , Rats , Rodentia/parasitology , Trypanosoma brucei rhodesiense/growth & development , Trypanosoma brucei rhodesiense/metabolism , Trypanosomiasis, African/blood , Trypanosomiasis, African/cerebrospinal fluid
5.
FEBS J ; 283(10): 1846-62, 2016 05.
Article in English | MEDLINE | ID: mdl-26945671

ABSTRACT

Trypanosomes that cause sleeping sickness endocytose apolipoprotein L1 (APOL1)-containing trypanolytic factors from human serum, leading to trypanolytic death through generation of APOL1-associated lytic pores in trypanosomal membranes. The trypanosome Trypanosoma brucei rhodesiense counteracts trypanolysis by expressing the surface protein serum response-associated (SRA), which can bind APOL1 common variant G0 to block its trypanolytic activity. However, two missense variants in the C terminal predicted coiled-coil (CC) domains of human APOL1 G1 (S342G/I384M) and G2 (ΔN388Y389) decrease or abrogate APOL1 binding to T. brucei rhodesiense SRA, thus preserving APOL1 trypanolytic activity. These evolutionarily selected APOL1 missense variants, found at a high frequency in some populations of African descent, also confer elevated risk of kidney disease. Understanding the SRA-APOL1 interaction and the role of APOL1 G1 and G2 variants in kidney disease demands structural characterization of the APOL1 CC domain. Using CD, heteronuclear NMR, and molecular dynamics (MD) simulation on structural homology models, we report here unique and dynamic solution conformations of nephropathy variants G1 and G2 as compared with the common variant G0. Conformational plasticity in G1 and G2 CC domains led to interhelical α1-α2 approximation coupled with secondary structural changes and delimited motional properties absent in the G0 CC domain. The G1 substitutions conferred local structural changes principally along helix α1, whereas the G2 deletion altered the structure of both helix α2 and helix α1. These dynamic features of APOL1 CC variants likely reflect their intrinsic structural properties, and should help interpret future APOL1 structural studies and define the contribution of APOL1 risk variants to kidney disease.


Subject(s)
Apolipoproteins/chemistry , Kidney Diseases/metabolism , Lipoproteins, HDL/chemistry , Mutation, Missense , Amino Acid Sequence , Animals , Apolipoprotein L1 , Apolipoproteins/genetics , Apolipoproteins/metabolism , Circular Dichroism , Cloning, Molecular , Humans , Lipoproteins, HDL/genetics , Lipoproteins, HDL/metabolism , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trypanosoma brucei rhodesiense/metabolism
6.
Cell ; 164(1-2): 246-257, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26771494

ABSTRACT

Intercellular communication between parasites and with host cells provides mechanisms for parasite development, immune evasion, and disease pathology. Bloodstream African trypanosomes produce membranous nanotubes that originate from the flagellar membrane and disassociate into free extracellular vesicles (EVs). Trypanosome EVs contain several flagellar proteins that contribute to virulence, and Trypanosoma brucei rhodesiense EVs contain the serum resistance-associated protein (SRA) necessary for human infectivity. T. b. rhodesiense EVs transfer SRA to non-human infectious trypanosomes, allowing evasion of human innate immunity. Trypanosome EVs can also fuse with mammalian erythrocytes, resulting in rapid erythrocyte clearance and anemia. These data indicate that trypanosome EVs are organelles mediating non-hereditary virulence factor transfer and causing host erythrocyte remodeling, inducing anemia.


Subject(s)
Extracellular Vesicles/metabolism , Membrane Glycoproteins/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei rhodesiense/cytology , Trypanosoma brucei rhodesiense/immunology , Trypanosomiasis, African/pathology , Trypanosomiasis, African/parasitology , Virulence Factors/metabolism , Anemia/pathology , Animals , Erythrocytes/parasitology , Flagella/metabolism , Humans , Immune Evasion , Mice , Proteome/metabolism , Rhodamines/analysis , Trypanosoma brucei rhodesiense/metabolism , Trypanosoma brucei rhodesiense/pathogenicity
7.
Bioorg Med Chem Lett ; 25(20): 4509-12, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26342866

ABSTRACT

Rhodesain, the major cathepsin L-like cysteine protease in the protozoan Trypanosoma brucei rhodesiense, the causative agent of African sleeping sickness, is a well-validated drug target. In this work, we used a fragment-based approach to identify inhibitors of this cysteine protease, and identified inhibitors of T. brucei. To discover inhibitors active against rhodesain and T. brucei, we screened a library of covalent fragments against rhodesain and conducted preliminary SAR studies. We envision that in vitro enzymatic assays will further expand the use of the covalent tethering method, a simple fragment-based drug discovery technique to discover covalent drug leads.


Subject(s)
Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cysteine/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei rhodesiense/metabolism , Cysteine/analogs & derivatives , Cysteine/chemistry , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/chemistry , Dose-Response Relationship, Drug , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/chemistry , Trypanosoma brucei brucei/enzymology
8.
Nat Commun ; 6: 8078, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26307671

ABSTRACT

Humans resist infection by the African parasite Trypanosoma brucei owing to the trypanolytic activity of the serum apolipoprotein L1 (APOL1). Following uptake by endocytosis in the parasite, APOL1 forms pores in endolysosomal membranes and triggers lysosome swelling. Here we show that APOL1 induces both lysosomal and mitochondrial membrane permeabilization (LMP and MMP). Trypanolysis coincides with MMP and consecutive release of the mitochondrial TbEndoG endonuclease to the nucleus. APOL1 is associated with the kinesin TbKIFC1, of which both the motor and vesicular trafficking VHS domains are required for MMP, but not for LMP. The presence of APOL1 in the mitochondrion is accompanied by mitochondrial membrane fenestration, which can be mimicked by knockdown of a mitochondrial mitofusin-like protein (TbMFNL). The BH3-like peptide of APOL1 is required for LMP, MMP and trypanolysis. Thus, trypanolysis by APOL1 is linked to apoptosis-like MMP occurring together with TbKIFC1-mediated transport of APOL1 from endolysosomal membranes to the mitochondrion.


Subject(s)
Apolipoproteins/metabolism , Kinesins/metabolism , Lipoproteins, HDL/metabolism , Lysosomes/metabolism , Mitochondrial Membranes/metabolism , Protozoan Proteins/metabolism , Apolipoprotein L1 , Apoptosis , Biological Transport , Endocytosis , Humans , Intracellular Membranes/metabolism , Permeability , Trypanosoma brucei brucei/metabolism , Trypanosoma brucei brucei/pathogenicity , Trypanosoma brucei gambiense/metabolism , Trypanosoma brucei gambiense/pathogenicity , Trypanosoma brucei rhodesiense/metabolism , Trypanosoma brucei rhodesiense/pathogenicity
9.
Kidney Int ; 88(4): 754-63, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25993319

ABSTRACT

A third of African Americans with sporadic focal segmental glomerulosclerosis (FSGS) or HIV-associated nephropathy (HIVAN) do not carry APOL1 renal risk genotypes. This raises the possibility that other APOL1 variants may contribute to kidney disease. To address this question, we sequenced all APOL1 exons in 1437 Americans of African and European descent, including 464 patients with biopsy-proven FSGS/HIVAN. Testing for association with 33 common and rare variants with FSGS/HIVAN revealed no association independent of strong recessive G1 and G2 effects. Seeking additional variants that might have been under selection by pathogens and could represent candidates for kidney disease risk, we also sequenced an additional 1112 individuals representing 53 global populations. Except for G1 and G2, none of the 7 common codon-altering variants showed evidence of selection or could restore lysis against trypanosomes causing human African trypanosomiasis. Thus, only APOL1 G1 and G2 confer renal risk, and other common and rare APOL1 missense variants, including the archaic G3 haplotype, do not contribute to sporadic FSGS and HIVAN in the US population. Hence, in most potential clinical or screening applications, our study suggests that sequencing APOL1 exons is unlikely to bring additional information compared to genotyping only APOL1 G1 and G2 risk alleles.


Subject(s)
AIDS-Associated Nephropathy/genetics , Apolipoproteins/genetics , Glomerulosclerosis, Focal Segmental/genetics , Lipoproteins, HDL/genetics , Polymorphism, Single Nucleotide , AIDS-Associated Nephropathy/diagnosis , AIDS-Associated Nephropathy/ethnology , Black or African American/genetics , Apolipoprotein L1 , Apolipoproteins/blood , Biopsy , Case-Control Studies , Exons , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Glomerulosclerosis, Focal Segmental/diagnosis , Glomerulosclerosis, Focal Segmental/ethnology , Haplotypes , Host-Parasite Interactions , Humans , Lipoproteins, HDL/blood , Male , Phenotype , Risk Assessment , Risk Factors , Sequence Analysis, DNA , Trypanosoma brucei gambiense/metabolism , Trypanosoma brucei gambiense/pathogenicity , Trypanosoma brucei rhodesiense/metabolism , Trypanosoma brucei rhodesiense/pathogenicity , United States/epidemiology , White People/genetics
10.
Nat Rev Microbiol ; 12(8): 575-84, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24975321

ABSTRACT

Humans can survive bloodstream infection by African trypanosomes, owing to the activity of serum complexes that have efficient trypanosome-killing ability. The two trypanosome subspecies that are responsible for human sleeping sickness--Trypanosoma brucei rhodesiense and Trypanosoma brucei gambiense--can evade this defence mechanism by expressing distinct resistance proteins. In turn, sequence variation in the gene that encodes the trypanosome-killing component in human serum has enabled populations in western Africa to restore resistance to T. b. rhodesiense, at the expense of the high probability of developing kidney sclerosis. These findings highlight the importance of resistance to trypanosomes in human evolution.


Subject(s)
Trypanosoma brucei gambiense/immunology , Trypanosoma brucei rhodesiense/immunology , Trypanosomiasis, African/immunology , Animals , Apolipoprotein L1 , Apolipoproteins/genetics , Apolipoproteins/metabolism , Biological Evolution , Disease Resistance/immunology , Humans , Lipoproteins, HDL/genetics , Lipoproteins, HDL/metabolism , Trypanosoma brucei gambiense/metabolism , Trypanosoma brucei rhodesiense/metabolism , Trypanosomiasis, African/genetics , Trypanosomiasis, African/metabolism
11.
Biomed Res Int ; 2014: 893272, 2014.
Article in English | MEDLINE | ID: mdl-24800253

ABSTRACT

The microtubule cytoskeleton forms the most prominent structural system in Trypanosoma brucei, undergoing extensive modifications during the cell cycle. Visualization of tyrosinated microtubules leads to a semiconservative mode of inheritance, whereas recent studies employing microtubule plus end tracking proteins have hinted at an asymmetric pattern of cytoskeletal inheritance. To further the knowledge of microtubule synthesis and inheritance during T. brucei cell cycle, the dynamics of the microtubule cytoskeleton was visualized by inducible YFP-α-tubulin expression. During new flagellum/flagellum attachment zone (FAZ) biogenesis and cell growth, YFP-α-tubulin was incorporated mainly between the old and new flagellum/FAZ complexes. Cytoskeletal modifications at the posterior end of the cells were observed with EB1, a microtubule plus end binding protein, particularly during mitosis. Additionally, the newly formed microtubules segregated asymmetrically, with the daughter cell inheriting the new flagellum/FAZ complex retaining most of the new microtubules. Together, our results suggest an intimate connection between new microtubule formation and new FAZ assembly, consequently leading to asymmetric microtubule inheritance and cell division.


Subject(s)
Luminescent Proteins/metabolism , Microtubules/metabolism , Recombinant Fusion Proteins/metabolism , Trypanosoma brucei rhodesiense/genetics , Trypanosoma brucei rhodesiense/metabolism , Tubulin/metabolism , Cell Line , Genes, Reporter/genetics , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Microtubules/chemistry , Microtubules/genetics , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Tubulin/analysis , Tubulin/genetics
12.
Parasitology ; 141(1): 28-36, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23931634

ABSTRACT

Target-based approaches for human African trypanosomiasis (HAT) and related parasites can be a valuable route for drug discovery for these diseases. However, care needs to be taken in selection of both the actual drug target and the chemical matter that is developed. In this article, potential criteria to aid target selection are described. Then the physiochemical properties of typical oral drugs are discussed and compared to those of known anti-parasitics.


Subject(s)
Drug Delivery Systems , Drug Discovery , Trypanocidal Agents/pharmacokinetics , Trypanosoma brucei gambiense/drug effects , Trypanosoma brucei rhodesiense/drug effects , Trypanosomiasis, African/drug therapy , Biological Availability , Dosage Forms , Drug Administration Routes , Drug Administration Schedule , Drug Resistance , Humans , Structure-Activity Relationship , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei gambiense/growth & development , Trypanosoma brucei gambiense/metabolism , Trypanosoma brucei rhodesiense/growth & development , Trypanosoma brucei rhodesiense/metabolism , Trypanosomiasis, African/parasitology
13.
PLoS Pathog ; 9(4): e1003317, 2013.
Article in English | MEDLINE | ID: mdl-23637606

ABSTRACT

Critical to human innate immunity against African trypanosomes is a minor subclass of human high-density lipoproteins, termed Trypanosome Lytic Factor-1 (TLF-1). This primate-specific molecule binds to a haptoglobin-hemoglobin receptor (HpHbR) on the surface of susceptible trypanosomes, initiating a lytic pathway. Group 1 Trypanosoma brucei gambiense causes human African Trypanosomiasis (HAT), escaping TLF-1 killing due to reduced uptake. Previously, we found that group 1 T. b. gambiense HpHbR (TbgHpHbR) mRNA levels were greatly reduced and the gene contained substitutions within the open reading frame. Here we show that a single, highly conserved amino acid in the TbgHpHbR ablates high affinity TLF-1 binding and subsequent endocytosis, thus evading TLF-1 killing. In addition, we show that over-expression of TbgHpHbR failed to rescue TLF-1 susceptibility. These findings suggest that the single substitution present in the TbgHpHbR directly contributes to the reduced uptake and resistance to TLF-1 seen in these important human pathogens.


Subject(s)
Haptoglobins/genetics , Haptoglobins/metabolism , Hemoglobins/genetics , Hemoglobins/metabolism , Lipoproteins, HDL/metabolism , Trypanosoma brucei gambiense/immunology , Trypanosoma brucei rhodesiense/immunology , Amino Acid Substitution , Cell Line , Endocytosis , Gene Knockout Techniques , Haptoglobins/chemistry , Hemoglobins/chemistry , Humans , Sequence Alignment , Trypanosoma brucei gambiense/chemistry , Trypanosoma brucei gambiense/genetics , Trypanosoma brucei gambiense/metabolism , Trypanosoma brucei rhodesiense/genetics , Trypanosoma brucei rhodesiense/metabolism , Trypanosomiasis, African/immunology
14.
PLoS Negl Trop Dis ; 6(6): e1668, 2012.
Article in English | MEDLINE | ID: mdl-22679520

ABSTRACT

Isothermal microcalorimetry is an established tool to measure heat flow of physical, chemical or biological processes. The metabolism of viable cells produces heat, and if sufficient cells are present, their heat production can be assessed by this method. In this study, we investigated the heat flow of two medically important protozoans, Trypanosoma brucei rhodesiense and Plasmodium falciparum. Heat flow signals obtained for these pathogens allowed us to monitor parasite growth on a real-time basis as the signals correlated with the number of viable cells. To showcase the potential of microcalorimetry for measuring drug action on pathogenic organisms, we tested the method with three antitrypanosomal drugs, melarsoprol, suramin and pentamidine and three antiplasmodial drugs, chloroquine, artemether and dihydroartemisinin, each at two concentrations on the respective parasite. With the real time measurement, inhibition was observed immediately by a reduced heat flow compared to that in untreated control samples. The onset of drug action, the degree of inhibition and the time to death of the parasite culture could conveniently be monitored over several days. Microcalorimetry is a valuable element to be added to the toolbox for drug discovery for protozoal diseases such as human African trypanosomiasis and malaria. The method could probably be adapted to other protozoan parasites, especially those growing extracellularly.


Subject(s)
Antiprotozoal Agents/pharmacology , Calorimetry/methods , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Trypanosoma brucei rhodesiense/drug effects , Trypanosoma brucei rhodesiense/growth & development , Cell Survival/drug effects , Humans , Parasitic Sensitivity Tests , Plasmodium falciparum/metabolism , Trypanosoma brucei rhodesiense/metabolism
15.
Eukaryot Cell ; 10(8): 1023-33, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21705681

ABSTRACT

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.


Subject(s)
Endosomes/metabolism , Membrane Glycoproteins/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei rhodesiense/physiology , Trypanosomiasis, African/parasitology , Cells, Cultured , Flagella/metabolism , Humans , Immune Evasion , Lipoproteins, HDL/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport , Trypanosoma brucei rhodesiense/metabolism , Trypanosomiasis, African/immunology
16.
Science ; 329(5993): 841-5, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20647424

ABSTRACT

African Americans have higher rates of kidney disease than European Americans. Here, we show that, in African Americans, focal segmental glomerulosclerosis (FSGS) and hypertension-attributed end-stage kidney disease (H-ESKD) are associated with two independent sequence variants in the APOL1 gene on chromosome 22 {FSGS odds ratio = 10.5 [95% confidence interval (CI) 6.0 to 18.4]; H-ESKD odds ratio = 7.3 (95% CI 5.6 to 9.5)}. The two APOL1 variants are common in African chromosomes but absent from European chromosomes, and both reside within haplotypes that harbor signatures of positive selection. ApoL1 (apolipoprotein L-1) is a serum factor that lyses trypanosomes. In vitro assays revealed that only the kidney disease-associated ApoL1 variants lysed Trypanosoma brucei rhodesiense. We speculate that evolution of a critical survival factor in Africa may have contributed to the high rates of renal disease in African Americans.


Subject(s)
Apolipoproteins/genetics , Black or African American/genetics , Glomerulosclerosis, Focal Segmental/genetics , Kidney Failure, Chronic/genetics , Lipoproteins, HDL/genetics , Polymorphism, Single Nucleotide , Trypanosoma brucei rhodesiense/metabolism , Africa , Alleles , Apolipoprotein L1 , Apolipoproteins/blood , Apolipoproteins/metabolism , Case-Control Studies , Cohort Studies , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Glomerulosclerosis, Focal Segmental/ethnology , Haplotypes , Humans , Hypertension/complications , Immunity, Innate , Kidney Failure, Chronic/ethnology , Kidney Failure, Chronic/etiology , Linkage Disequilibrium , Lipoproteins, HDL/blood , Lipoproteins, HDL/metabolism , Logistic Models , Molecular Motor Proteins/genetics , Myosin Heavy Chains/genetics , Recombinant Proteins/metabolism , Selection, Genetic , Sequence Deletion , Trypanosomiasis, African/genetics , Trypanosomiasis, African/parasitology
17.
PLoS One ; 5(3): e9630, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20224787

ABSTRACT

BACKGROUND: The only available diagnostic method for East African trypanosomiasis is light microscopy of blood samples. A simple immunodiagnostic would greatly aid trypanosomiasis control. METHODOLOGY AND PRINCIPAL FINDINGS: To find trypanosome proteins that are specifically recognised by sera from human sleeping sickness patients, we have screened the Trypanosoma brucei brucei proteome by Western blotting. Using cytosolic, cytoskeletal and glycosomal fractions, we found that the vast majority of abundant trypanosome proteins is not specifically recognised by patient sera. We identified phosphoglycerate kinase (PGKC), heat shock protein (HSP70), and histones H2B and H3 as possible candidate diagnostic antigens. These proteins, plus paraflagellar rod protein 1, rhodesain (a cysteine protease), and an extracellular fragment of the Trypanosoma brucei nucleoside transporter TbNT10, were expressed in E. coli and tested for reactivity with patient and control sera. Only TbHSP70 was preferentially recognized by patient sera, but the sensitivity and specificity were insufficient for use of TbHSP70 alone as a diagnostic. Immunoprecipitation using a native protein extract revealed no specifically reacting proteins. CONCLUSIONS: No abundant T. brucei soluble, glycosomal or cytoskeletal protein is likely to be useful in diagnosis. To find useful diagnostic antigens it will therefore be necessary to use more sophisticated proteomic methods, or to test a very large panel of candidate proteins.


Subject(s)
Trypanosoma brucei rhodesiense/genetics , Trypanosoma brucei rhodesiense/metabolism , Trypanosomiasis/diagnosis , Trypanosomiasis/parasitology , Cloning, Molecular , HSP70 Heat-Shock Proteins/biosynthesis , Histones/biosynthesis , Humans , Phosphoglycerate Kinase/biosynthesis , Predictive Value of Tests , Proteomics/methods , Reagent Kits, Diagnostic , Sensitivity and Specificity , Serologic Tests , Subcellular Fractions , Trypanosoma brucei rhodesiense/chemistry , Trypanosomiasis/blood
18.
DNA Cell Biol ; 29(5): 215-27, 2010 May.
Article in English | MEDLINE | ID: mdl-20307190

ABSTRACT

The potential association of variant surface glycoprotein (VSG) gene expression with clonal expression of virulence in African trypanosomes was addressed. Two populations of clonally related trypanosomes, which differ dramatically in virulence for the infected host, but display the same apparent VSG surface coat phenotype, were characterized with respect to the VSG genes expressed as well as the chromosome telomeric expression sites (ES) utilized for VSG gene transcription. The VSG gene sequences expressed by clones LouTat 1 and LouTat 1A of Trypanosoma brucei rhodesiense were identical, and gene expression in both clones occurred precisely by the same gene conversion events (duplication and transposition), which generated an expression-linked copy (ELC) of the VSG gene. The ELC was present on the same genomic restriction fragments in both populations and resided in the telomere of a 330-kb chromosome; a single basic copy of the LouTat 1/1A VSG gene, present in all variants of the LouTat 1 serodeme, was located at an internal site of a 1.5-Mb chromosome. Restriction endonuclease mapping of the ES telomere revealed that the VSG ELC of clones LouTat 1 and 1A resides in the same site. Therefore, these findings provide evidence that the VSG gene ES and, potentially, any cotranscribed ES-associated genes do not play a role in the clonal regulation of virulence because trypanosome clones LouTat 1 and 1A, which differ markedly in their virulence properties, both express identical VSG genes from the same chromosome telomeric ES.


Subject(s)
Genes, Protozoan , Trypanosoma brucei rhodesiense/metabolism , Variant Surface Glycoproteins, Trypanosoma/metabolism , Amino Acid Sequence , Animals , Base Sequence , Mice , Molecular Sequence Data , Telomere/metabolism , Trypanosoma brucei rhodesiense/genetics , Trypanosoma brucei rhodesiense/pathogenicity , Variant Surface Glycoproteins, Trypanosoma/genetics , Variant Surface Glycoproteins, Trypanosoma/immunology
19.
PLoS Pathog ; 5(12): e1000685, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19997494

ABSTRACT

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.


Subject(s)
Apolipoproteins/physiology , Cell Survival/drug effects , Lipoproteins, HDL/physiology , Membrane Glycoproteins/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/physiology , Trypanosoma brucei rhodesiense/physiology , Amino Acid Sequence , Animals , Apolipoprotein L1 , Apolipoproteins/genetics , Apolipoproteins/metabolism , Apolipoproteins/pharmacology , DNA Mutational Analysis , Humans , Leucine Zippers/genetics , Lipoproteins, HDL/genetics , Lipoproteins, HDL/metabolism , Lipoproteins, HDL/pharmacology , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Papio anubis , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Pore Forming Cytotoxic Proteins/pharmacology , Protein Binding , Sequence Alignment , Thermodynamics , Trypanocidal Agents/metabolism , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/metabolism , Trypanosoma brucei rhodesiense/metabolism
20.
Proc Natl Acad Sci U S A ; 106(42): 17933-8, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19815526

ABSTRACT

Trypanosoma brucei is one of the most ancient eukaryotes where RNA interference (RNAi) is operational and is the only single-cell pathogen where RNAi has been extensively studied and used as a tool for functional analyses. Here, we report that the T. brucei RNAi pathway, although relying on a single Argonaute protein (AGO1), is initiated by the activities of two distinct Dicer-like enzymes. Both TbDCL1, a mostly cytoplasmic protein, and the previously undescribed nuclear enzyme TbDCL2 contribute to the biogenesis of siRNAs from retroposons. However, TbDCL2 has a predominant role in generating siRNAs from chromosomal internal repeat transcripts that accumulate at the nucleolus in RNAi-deficient cells and in initiating the endogenous RNAi response against retroposons and repeats alike. Moreover, siRNAs generated by both TbDCL1 and TbDCL2 carry a 5'-monophosphate and a blocked 3' terminus, suggesting that 3' end modification is an ancient trait of siRNAs. We thus propose a model whereby TbDCL2 fuels the T. brucei nuclear RNAi pathway and TbDCL1 patrols the cytoplasm, posttranscriptionally silencing potentially harmful nucleic acid parasites that may access the cytoplasm. Nevertheless, we also provide evidence for cross-talk between the two Dicer-like enzymes, because TbDCL2 is implicated in the generation of 35- to 65-nucleotide intermediate transcripts that appear to be substrates for TbDCL1. Our finding that dcl2KO cells are more sensitive to RNAi triggers than wild-type cells has significant implications for reverse genetic analyses in this important human pathogen.


Subject(s)
Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA Interference , Ribonuclease III/genetics , Ribonuclease III/metabolism , Trypanosoma brucei rhodesiense/genetics , Trypanosoma brucei rhodesiense/metabolism , Animals , Humans , RNA, Protozoan/genetics , RNA, Small Interfering/genetics , Retroelements/genetics , Transcription, Genetic , Trypanosoma brucei rhodesiense/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...