Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
1.
RNA ; 29(2): 228-240, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36400448

RESUMEN

Mitochondrial gene expression in trypanosomes requires numerous multiprotein complexes that are unique to kinetoplastids. Among these, the most well characterized are RNA editing catalytic complexes (RECCs) that catalyze the guide RNA (gRNA)-specified insertion and deletion of uridines during mitochondrial mRNA maturation. This post-transcriptional resequencing of mitochondrial mRNAs can be extensive, involving dozens of different gRNAs and hundreds of editing sites with most of the mature mRNA sequences resulting from the editing process. Proper coordination of the editing with the cognate gRNAs is attributed to RNA editing substrate-binding complexes (RESCs), which are also required for RNA editing. Although the precise mechanism of RESC function is less well understood, their affinity for binding both editing substrates and products suggests that these complexes may provide a scaffold for RECC catalytic processing. KRGG1 has been shown to bind RNAs, and although affinity purification co-isolates RESC complexes, its role in RNA editing remains uncertain. We show here that KRGG1 is essential in BF parasites and required for normal editing. KRGG1 repression results in reduced amounts of edited A6 mRNA and increased amounts of edited ND8 mRNA. Sequence and structure analysis of KRGG1 identified a region of homology with RESC6, and both proteins have predicted tandem helical repeats that resemble ARM/HEAT motifs. The ARM/HEAT-like region is critical for function as exclusive expression of mutated KRGG1 results in growth inhibition and disruption of KRGG1 association with RESCs. These results indicate that KRGG1 is critical for RNA editing and its specific function is associated with RESC activity.


Asunto(s)
Edición de ARN , Trypanosoma brucei brucei , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/metabolismo , ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Guía de Kinetoplastida/genética , ARN Protozoario/genética , ARN Protozoario/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
2.
RNA ; 29(10): 1591-1609, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37474258

RESUMEN

The gRNA directed U-insertion and deletion editing of mitochondrial mRNAs that is essential in different life-cycle stages for the protozoan parasite Trypanosoma brucei is performed by three similar multiprotein catalytic complexes (CCs) that contain the requisite enzymes. These CCs also contain a common set of eight proteins that have no apparent direct catalytic function, including six that have an OB-fold domain. We show here that one of these OB-fold proteins, KREPA3 (A3), has structural homology to other editing proteins, is essential for editing, and is multifunctional. We investigated A3 function by analyzing the effects of single amino acid loss of function mutations, most of which were identified by screening bloodstream form (BF) parasites for loss of growth following random mutagenesis. Mutations in the zinc fingers (ZFs), an intrinsically disordered region (IDR), and several within or near the carboxy-terminal OB-fold domain variably impacted CC structural integrity and editing. Some mutations resulted in almost complete loss of CCs and its proteins and editing, whereas others retained CCs but had aberrant editing. All but a mutation which is near the OB-fold affected growth and editing in BF but not procyclic form (PF) parasites. These data indicate that multiple positions within A3 have essential functions that contribute to the structural integrity of CCs, the precision of editing and the developmental differences in editing between BF and PF stages.


Asunto(s)
ARN , Trypanosoma brucei brucei , ARN/genética , Trypanosoma brucei brucei/metabolismo , Edición de ARN , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Mutación , ARN Protozoario/genética , ARN Protozoario/metabolismo
3.
PLoS Pathog ; 19(5): e1011051, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37195999

RESUMEN

Understanding immune mechanisms that mediate malaria protection is critical for improving vaccine development. Vaccination with radiation-attenuated Plasmodium falciparum sporozoites (PfRAS) induces high level of sterilizing immunity against malaria and serves as a valuable tool for the study of protective mechanisms. To identify vaccine-induced and protection-associated responses during malarial infection, we performed transcriptome profiling of whole blood and in-depth cellular profiling of PBMCs from volunteers who received either PfRAS or noninfectious mosquito bites, followed by controlled human malaria infection (CHMI) challenge. In-depth single-cell profiling of cell subsets that respond to CHMI in mock-vaccinated individuals showed a predominantly inflammatory transcriptome response. Whole blood transcriptome analysis revealed that gene sets associated with type I and II interferon and NK cell responses were increased in prior to CHMI while T and B cell signatures were decreased as early as one day following CHMI in protected vaccinees. In contrast, non-protected vaccinees and mock-vaccinated individuals exhibited shared transcriptome changes after CHMI characterized by decreased innate cell signatures and inflammatory responses. Additionally, immunophenotyping data showed different induction profiles of vδ2+ γδ T cells, CD56+ CD8+ T effector memory (Tem) cells, and non-classical monocytes between protected vaccinees and individuals developing blood-stage parasitemia, following treatment and resolution of infection. Our data provide key insights in understanding immune mechanistic pathways of PfRAS-induced protection and infective CHMI. We demonstrate that vaccine-induced immune response is heterogenous between protected and non-protected vaccinees and that inducted-malaria protection by PfRAS is associated with early and rapid changes in interferon, NK cell and adaptive immune responses. Trial Registration: ClinicalTrials.gov NCT01994525.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum , Malaria , Humanos , Animales , Malaria Falciparum/prevención & control , Plasmodium falciparum/genética , Vacunación , Interferones , Inmunidad , Esporozoítos
4.
Nucleic Acids Res ; 51(13): 6944-6965, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37246647

RESUMEN

U-insertion/deletion (U-indel) RNA editing in trypanosome mitochondria is directed by guide RNAs (gRNAs). This editing may developmentally control respiration in bloodstream forms (BSF) and insect procyclic forms (PCF). Holo-editosomes include the accessory RNA Editing Substrate Binding Complex (RESC) and RNA Editing Helicase 2 Complex (REH2C), but the specific proteins controlling differential editing remain unknown. Also, RNA editing appears highly error prone because most U-indels do not match the canonical pattern. However, despite extensive non-canonical editing of unknown functions, accurate canonical editing is required for normal cell growth. In PCF, REH2C controls editing fidelity in RESC-bound mRNAs. Here, we report that KREH2, a REH2C-associated helicase, developmentally controls programmed non-canonical editing, including an abundant 3' element in ATPase subunit 6 (A6) mRNA. The 3' element sequence is directed by a proposed novel regulatory gRNA. In PCF, KREH2 RNAi-knockdown up-regulates the 3' element, which establishes a stable structure hindering element removal by canonical initiator-gRNA-directed editing. In BSF, KREH2-knockdown does not up-regulate the 3' element but reduces its high abundance. Thus, KREH2 differentially controls extensive non-canonical editing and associated RNA structure via a novel regulatory gRNA, potentially hijacking factors as a 'molecular sponge'. Furthermore, this gRNA is bifunctional, serving in canonical CR4 mRNA editing whilst installing a structural element in A6 mRNA.


Asunto(s)
Trypanosoma brucei brucei , Trypanosoma , ARN Mensajero/metabolismo , ARN Helicasas/genética , ARN Helicasas/metabolismo , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/metabolismo , Trypanosoma/genética , ARN/genética , ARN Protozoario/genética , ARN Protozoario/metabolismo
5.
PLoS Pathog ; 18(2): e1010282, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35108339

RESUMEN

Immunization with radiation-attenuated sporozoites (RAS) can confer sterilizing protection against malaria, although the mechanisms behind this protection are incompletely understood. We performed a systems biology analysis of samples from the Immunization by Mosquito with Radiation Attenuated Sporozoites (IMRAS) trial, which comprised P. falciparum RAS-immunized (PfRAS), malaria-naive participants whose protection from malaria infection was subsequently assessed by controlled human malaria infection (CHMI). Blood samples collected after initial PfRAS immunization were analyzed to compare immune responses between protected and non-protected volunteers leveraging integrative analysis of whole blood RNA-seq, high parameter flow cytometry, and single cell CITEseq of PBMCs. This analysis revealed differences in early innate immune responses indicating divergent paths associated with protection. In particular, elevated levels of inflammatory responses early after the initial immunization were detrimental for the development of protective adaptive immunity. Specifically, non-classical monocytes and early type I interferon responses induced within 1 day of PfRAS vaccination correlated with impaired immunity. Non-protected individuals also showed an increase in Th2 polarized T cell responses whereas we observed a trend towards increased Th1 and T-bet+ CD8 T cell responses in protected individuals. Temporal differences in genes associated with natural killer cells suggest an important role in immune regulation by these cells. These findings give insight into the immune responses that confer protection against malaria and may guide further malaria vaccine development. Trial registration: ClinicalTrials.gov NCT01994525.


Asunto(s)
Inmunidad , Inflamación , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Esporozoítos/inmunología , Adulto , Animales , Anopheles/parasitología , Femenino , Humanos , Inmunización/métodos , Mordeduras y Picaduras de Insectos/inmunología , Malaria Falciparum/parasitología , Masculino , Mosquitos Vectores/parasitología , Linfocitos T/inmunología , Vacunación/métodos , Vacunas Atenuadas/inmunología
6.
Nucleic Acids Res ; 50(17): 10123-10139, 2022 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-36095119

RESUMEN

Each of the three similar RNA Editing Catalytic Complexes (RECCs) that perform gRNA-directed uridine insertion and deletion during Trypanosoma brucei mitochondrial (mt) mRNA editing has a distinct endonuclease activity that requires two related RNase III proteins, with only one competent for catalysis. We identified multiple loss-of-function mutations in the RNase III and other motifs of the non-catalytic KREPB6, KREPB7, and KREPB8 components by random mutagenesis and screening. These mutations had various effects on growth, editing, and both the abundances and RECC associations of these RNase III protein pairs in bloodstream form (BF) and procyclic form (PF) cells. Protein structure modelling predicted that the Zinc Finger (ZnF) of each paired RNase III protein contacts RNA positioned at the heterodimeric active site which is flanked by helices of a novel RNase III-Associated Motif (RAM). The results indicate that the protein domains of the non-catalytic subunits function together in RECC integrity, substrate binding, and editing site recognition during the multistep RNA editing process. Additionally, several mutants display distinct functional consequences in different life cycle stages. These results highlight the complementary roles of protein pairs and three RECCs within the complicated T. brucei mRNA editing machinery that matures mt mRNAs differentially between developmental stages.


Asunto(s)
Proteínas Protozoarias/metabolismo , Ribonucleasa III/metabolismo , Trypanosoma brucei brucei , Endonucleasas/genética , Endonucleasas/metabolismo , ARN/metabolismo , Edición de ARN , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Protozoario/genética , ARN Protozoario/metabolismo , Trypanosoma brucei brucei/metabolismo , Uridina/metabolismo
7.
RNA ; 25(9): 1150-1163, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31171708

RESUMEN

Multiprotein editosomes catalyze gRNA-specified insertion and deletion of uridines to create functional mitochondrial mRNAs in Trypanosoma brucei Three functionally distinct editosomes are distinguished by their single KREN1, KREN2, or KREN3 RNase III endonuclease and, respectively, KREPB8, KREPB7, and KREPB6 partner proteins. These endonucleases perform the first catalytic step of editing, cleaving mRNA in diverse mRNA/gRNA heteroduplex substrates. We identified divergent and likely noncatalytic RNase III domains in KREPB4, KREPB5, KREPB6, KREPB7, KREPB8, KREPB9, and KREPB10 editosome proteins. Because known RNase III endonuclease functional domains are dimeric, the editing endonucleases may form heterodimers with one or more of these divergent RNase III proteins. We show here using conditional null cell lines that KREPB6, KREPB7, and KREPB8 are essential in both procyclic form (PF) and bloodstream (BF) cells. Loss of these proteins results in growth defects and loss of editing in vivo, as does mutation of their RNase III domain that is predicted to prevent dimerization. Loss of KREPB6, KREPB7, or KREPB8 also dramatically reduces cognate endonuclease abundance, as does the RNase III mutation, indicating that RNase III interactions with their partner proteins stabilize the endonucleases. The phenotypic consequences of repression are more severe in BF than in PF, indicating differences in endonuclease function between developmental stages that could impact regulation of editing. These results suggest that KREPB6, KREPB7, and KREPB8 form heterodimers with their respective endonucleases to perform mRNA cleavage. We also present a model wherein editosome proteins with divergent RNase III domains function in substrate selection via enzyme-pseudoenzyme interactions.


Asunto(s)
Proteínas Protozoarias/genética , Edición de ARN/genética , Ribonucleasa III/genética , Trypanosoma brucei brucei/genética , Animales , Línea Celular , Endonucleasas/genética , Mutación/genética , ARN Guía de Kinetoplastida/genética , ARN Mensajero/genética , ARN Mitocondrial/genética , ARN Protozoario/genética , Uridina/genética
8.
Virol J ; 18(1): 28, 2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33499880

RESUMEN

BACKGROUND: Diverse vaccination outcomes and protection levels among different populations pose a serious challenge to the development of an effective malaria vaccine. Co-infections are among many factors associated with immune dysfunction and sub-optimal vaccination outcomes. Chronic, asymptomatic viral infections can contribute to the modulation of vaccine efficacy through various mechanisms. Human Pegivirus-1 (HPgV-1) persists in immune cells thereby potentially modulating immune responses. We investigated whether Pegivirus infection influences vaccine-induced responses and protection in African volunteers undergoing whole P. falciparum sporozoites-based malaria vaccination and controlled human malaria infections (CHMI). METHODS: HPgV-1 prevalence was quantified by RT-qPCR in plasma samples of 96 individuals before, post vaccination with PfSPZ Vaccine and after CHMI in cohorts from Tanzania and Equatorial Guinea. The impact of HPgV-1 infection was evaluated on (1) systemic cytokine and chemokine levels measured by Luminex, (2) PfCSP-specific antibody titers quantified by ELISA, (3) asexual blood-stage parasitemia pre-patent periods and parasite multiplication rates, (4) HPgV-1 RNA levels upon asexual blood-stage parasitemia induced by CHMI. RESULTS: The prevalence of HPgV-1 was 29.2% (28/96) and sequence analysis of the 5' UTR and E2 regions revealed the predominance of genotypes 1, 2 and 5. HPgV-1 infection was associated with elevated systemic levels of IL-2 and IL-17A. Comparable vaccine-induced anti-PfCSP antibody titers, asexual blood-stage multiplication rates and pre-patent periods were observed in HPgV-1 positive and negative individuals. However, a tendency for higher protection levels was detected in the HPgV-1 positive group (62.5%) compared to the negative one (51.6%) following CHMI. HPgV-1 viremia levels were not significantly altered after CHMI. CONCLUSIONS: HPgV-1 infection did not alter PfSPZ Vaccine elicited levels of PfCSP-specific antibody responses and parasite multiplication rates. Ongoing HPgV-1 infection appears to improve to some degree protection against CHMI in PfSPZ-vaccinated individuals. This is likely through modulation of immune system activation and systemic cytokines as higher levels of IL-2 and IL17A were observed in HPgV-1 infected individuals. CHMI is safe and well tolerated in HPgV-1 infected individuals. Identification of cell types and mechanisms of both silent and productive infection in individuals will help to unravel the biology of this widely present but largely under-researched virus.


Asunto(s)
Coinfección/inmunología , Infecciones por Flaviviridae/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Esporozoítos/inmunología , Adolescente , Adulto , Estudios de Cohortes , Coinfección/complicaciones , Coinfección/parasitología , Coinfección/virología , Femenino , Infecciones por Flaviviridae/sangre , Infecciones por Flaviviridae/complicaciones , Infecciones por Flaviviridae/epidemiología , Guinea , Humanos , Vacunas contra la Malaria/administración & dosificación , Masculino , Persona de Mediana Edad , Pegivirus/genética , Pegivirus/inmunología , Plasmodium falciparum/inmunología , Ensayos Clínicos Controlados Aleatorios como Asunto , Tanzanía , Vacunación , Potencia de la Vacuna , Adulto Joven
9.
Cytometry A ; 97(10): 1019-1023, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32415811

RESUMEN

This 27-color flow cytometry panel was developed in order to assess immunological changes over the course of an immunization and challenge regimen in two experimental malaria vaccine trials. The aim of the study was to find correlates of vaccine-induced protection. Several studies have indicated that protection against malaria appears to involve immune responses at various immunological sites, with liver-resident responses playing an essential role. As it is not feasible to monitor the immune responses within the liver in humans, this panel is developed with the aim to thoroughly characterize the immune responses over time in blood in addition to detecting changes that might reflect what happens in other immunological sites like the liver. The focus of this panel is to detect several innate lymphoid cell populations, including NK cells and their activation status. Moreover, unconventional T cells like mucosal associated invariant T cells and γδ T cells are assessed in the panel. © 2020 International Society for Advancement of Cytometry.


Asunto(s)
Vacunas contra la Malaria , Células T Invariantes Asociadas a Mucosa , Citometría de Flujo , Humanos , Inmunidad Innata , Células Asesinas Naturales/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Subgrupos de Linfocitos T/inmunología
10.
RNA ; 23(11): 1672-1684, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28802260

RESUMEN

Uridine insertion and deletion RNA editing generates functional mitochondrial mRNAs in Trypanosoma brucei, and several transcripts are differentially edited in bloodstream (BF) and procyclic form (PF) cells correlating with changes in mitochondrial function. Editing is catalyzed by three ∼20S editosomes that have a common set of 12 proteins, but are typified by mutually exclusive RNase III KREN1, N2, and N3 endonucleases with distinct cleavage specificities. KREPB4 is a common editosome protein that has a degenerate RNase III domain lacking conserved catalytic residues, in addition to zinc-finger and Pumilio/fem-3 mRNA binding factor (PUF) motifs. Here we show that KREPB4 is essential for BF and PF growth, in vivo RNA editing, and editosome integrity, but that loss of KREPB4 has differential effects on editosome components and complexes between BF and PF cells. We used targeted mutagenesis to investigate the functions of the conserved PUF and RNase III domains in both life-cycle stages and show that the PUF motif is not essential for function in BF or PF. In contrast, specific mutations in the RNase III domain severely inhibit BF and PF growth and editing, and disrupt ∼20S editosomes, while others indicate that the RNase III domain is noncatalytic. We further show that KREPB4, specifically the noncatalytic RNase III domain, is required for the association of KREN1, N2, and N3 with PF editosomes. These results, combined with previous studies, support a model in which KREPB4 acts as a pseudoenzyme to form the noncatalytic half of an RNase III heterodimer with the editing endonucleases.


Asunto(s)
Proteínas Protozoarias/metabolismo , Edición de ARN , Proteínas de Unión al ARN/metabolismo , Trypanosoma brucei brucei/metabolismo , Endonucleasas/metabolismo , Técnicas de Silenciamiento del Gen , Genes Protozoarios , Modelos Biológicos , Mutación , Dominios Proteicos , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mitocondrial , ARN Protozoario/genética , ARN Protozoario/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Ribonucleasa III/química , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/crecimiento & desarrollo
11.
J Immunol ; 199(1): 107-118, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28576979

RESUMEN

Animal model studies highlight the role of innate-like lymphocyte populations in the early inflammatory response and subsequent parasite control following Plasmodium infection. IFN-γ production by these lymphocytes likely plays a key role in the early control of the parasite and disease severity. Analyzing human innate-like T cell and NK cell responses following infection with Plasmodium has been challenging because the early stages of infection are clinically silent. To overcome this limitation, we examined blood samples from a controlled human malaria infection (CHMI) study in a Tanzanian cohort, in which volunteers underwent CHMI with a low or high dose of Plasmodium falciparum sporozoites. The CHMI differentially affected NK, NKT (invariant NKT), and mucosal-associated invariant T cell populations in a dose-dependent manner, resulting in an altered composition of this innate-like lymphocyte compartment. Although these innate-like responses are typically thought of as short-lived, we found that changes persisted for months after the infection was cleared, leading to significantly increased frequencies of mucosal-associated invariant T cells 6 mo postinfection. We used single-cell RNA sequencing and TCR αß-chain usage analysis to define potential mechanisms for this expansion. These single-cell data suggest that this increase was mediated by homeostatic expansion-like mechanisms. Together, these data demonstrate that CHMI leads to previously unappreciated long-lasting alterations in the human innate-like lymphocyte compartment. We discuss the consequences of these changes for recurrent parasite infection and infection-associated pathologies and highlight the importance of considering host immunity and infection history for vaccine design.


Asunto(s)
Inmunidad Innata , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Malaria Falciparum/inmunología , Adulto , Interacciones Huésped-Patógeno , Humanos , Inmunidad Mucosa , Interferón gamma/inmunología , Vacunas contra la Malaria , Malaria Falciparum/parasitología , Masculino , Células T Invariantes Asociadas a Mucosa/inmunología , Parasitemia/inmunología , Plasmodium falciparum/inmunología , Plasmodium falciparum/fisiología , Esporozoítos/inmunología , Tanzanía , Factores de Tiempo , Adulto Joven
12.
Nucleic Acids Res ; 45(8): 4667-4686, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28334821

RESUMEN

RNA editing is an essential post-transcriptional process that creates functional mitochondrial mRNAs in Kinetoplastids. Multiprotein editosomes catalyze pre-mRNA cleavage, uridine (U) insertion or deletion, and ligation as specified by guide RNAs. Three functionally and compositionally distinct editosomes differ by the mutually exclusive presence of the KREN1, KREN2 or KREN3 endonuclease and their associated partner proteins. Because endonuclease cleavage is a likely point of regulation for RNA editing, we elucidated endonuclease specificity in vivo. We used a mutant gamma ATP synthase allele (MGA) to circumvent the normal essentiality of the editing endonucleases, and created cell lines in which both alleles of one, two or all three of the endonucleases were deleted. Cells lacking multiple endonucleases had altered editosome sedimentation on glycerol gradients and substantial defects in overall editing. Deep sequencing analysis of RNAs from such cells revealed clear discrimination by editosomes between sites of deletion versus insertion editing and preferential but overlapping specificity for sites of insertion editing. Thus, endonuclease specificities in vivo are distinct but with some functional overlap. The overlapping specificities likely accommodate the more numerous sites of insertion versus deletion editing as editosomes collaborate to accurately edit thousands of distinct editing sites in vivo.


Asunto(s)
Endonucleasas/genética , Proteínas Protozoarias/genética , Edición de ARN , ARN Mensajero/genética , ARN Protozoario/genética , Trypanosoma brucei brucei/genética , Secuencia de Bases , Endonucleasas/metabolismo , Eliminación de Gen , Glicerol/farmacología , Isoenzimas/genética , Isoenzimas/metabolismo , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Proteínas Protozoarias/metabolismo , División del ARN , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , ARN Mensajero/metabolismo , ARN Mitocondrial , ARN Protozoario/metabolismo , Alineación de Secuencia , Especificidad por Sustrato , Transfección , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/enzimología
13.
Proc Natl Acad Sci U S A ; 113(42): E6476-E6485, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27708162

RESUMEN

Uridine insertion and deletion RNA editing generates functional mitochondrial mRNAs in Trypanosoma brucei Editing is catalyzed by three distinct ∼20S editosomes that have a common set of 12 proteins, but are typified by mutually exclusive RNase III endonucleases with distinct cleavage specificities and unique partner proteins. Previous studies identified a network of protein-protein interactions among a subset of common editosome proteins, but interactions among the endonucleases and their partner proteins, and their interactions with common subunits were not identified. Here, chemical cross-linking and mass spectrometry, comparative structural modeling, and genetic and biochemical analyses were used to define the molecular architecture and subunit organization of purified editosomes. We identified intra- and interprotein cross-links for all editosome subunits that are fully consistent with editosome protein structures and previously identified interactions, which we validated by genetic and biochemical studies. The results were used to create a highly detailed map of editosome protein domain proximities, leading to identification of molecular interactions between subunits, insights into the functions of noncatalytic editosome proteins, and a global understanding of editosome architecture.


Asunto(s)
Proteínas Protozoarias/metabolismo , Edición de ARN , ARN Protozoario/genética , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/metabolismo , Biología Computacional/métodos , Endonucleasas/metabolismo , Modelos Moleculares , Complejos Multiproteicos/metabolismo , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Mapas de Interacción de Proteínas , Proteoma , Proteómica/métodos , Proteínas Protozoarias/química , Proteínas de Unión al ARN/metabolismo , Reproducibilidad de los Resultados
14.
J Biol Chem ; 290(41): 24914-31, 2015 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-26304125

RESUMEN

Uridine insertion and deletion RNA editing generates functional mitochondrial mRNAs in Trypanosoma brucei. The mRNAs are differentially edited in bloodstream form (BF) and procyclic form (PF) life cycle stages, and this correlates with the differential utilization of glycolysis and oxidative phosphorylation between the stages. The mechanism that controls this differential editing is unknown. Editing is catalyzed by multiprotein ∼20S editosomes that contain endonuclease, 3'-terminal uridylyltransferase, exonuclease, and ligase activities. These editosomes also contain KREPB5 and KREPA3 proteins, which have no functional catalytic motifs, but they are essential for parasite viability, editing, and editosome integrity in BF cells. We show here that repression of KREPB5 or KREPA3 is also lethal in PF, but the effects on editosome structure differ from those in BF. In addition, we found that point mutations in KREPB5 or KREPA3 differentially affect cell growth, editosome integrity, and RNA editing between BF and PF stages. These results indicate that the functions of KREPB5 and KREPA3 editosome proteins are adjusted between the life cycle stages. This implies that these proteins are involved in the processes that control differential editing and that the 20S editosomes differ between the life cycle stages.


Asunto(s)
Estadios del Ciclo de Vida , Proteínas Protozoarias/metabolismo , Edición de ARN , Trypanosoma brucei brucei/crecimiento & desarrollo , Trypanosoma brucei brucei/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sangre/parasitología , Línea Celular , Resistencia a Medicamentos/genética , Estadios del Ciclo de Vida/efectos de los fármacos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Estructura Terciaria de Proteína , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Ribonucleasa III/química , Ribonucleasa III/metabolismo , Tetraciclina/farmacología , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/metabolismo
15.
Mol Microbiol ; 97(5): 1006-20, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26043892

RESUMEN

The causative agent of human African trypanosomiasis, Trypanosoma brucei, lacks de novo purine biosynthesis and depends on purine salvage from the host. The purine salvage pathway is redundant and contains two routes to guanosine-5'-monophosphate (GMP) formation: conversion from xanthosine-5'-monophosphate (XMP) by GMP synthase (GMPS) or direct salvage of guanine by hypoxanthine-guanine phosphoribosyltransferase (HGPRT). We show recombinant T. brucei GMPS efficiently catalyzes GMP formation. Genetic knockout of GMPS in bloodstream parasites led to depletion of guanine nucleotide pools and was lethal. Growth of gmps null cells was only rescued by supraphysiological guanine concentrations (100 µM) or by expression of an extrachromosomal copy of GMPS. Hypoxanthine was a competitive inhibitor of guanine rescue, consistent with a common uptake/metabolic conversion mechanism. In mice, gmps null parasites were unable to establish an infection demonstrating that GMPS is essential for virulence and that plasma guanine is insufficient to support parasite purine requirements. These data validate GMPS as a potential therapeutic target for treatment of human African trypanosomiasis. The ability to strategically inhibit key metabolic enzymes in the purine pathway unexpectedly bypasses its functional redundancy by exploiting both the nature of pathway flux and the limited nutrient environment of the parasite's extracellular niche.


Asunto(s)
Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Purinas/metabolismo , Trypanosoma brucei brucei/enzimología , Trypanosoma brucei brucei/fisiología , Adenosina/metabolismo , Animales , Sitios de Unión , Ciclo Celular , Técnicas de Inactivación de Genes , Guanina/metabolismo , Guanina/farmacología , Guanosina Monofosfato/metabolismo , Humanos , Hipoxantina/metabolismo , Hipoxantina/farmacología , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/patogenicidad , Tripanosomiasis Africana/parasitología , Tripanosomiasis Africana/terapia
16.
Proc Natl Acad Sci U S A ; 110(14): 5392-7, 2013 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-23471987

RESUMEN

Malaria parasites use hemoglobin (Hb) as a major nutrient source in the intraerythrocytic stage, during which heme is converted to hemozoin (Hz). The formation of Hz is essential for parasite survival, but to date, the underlying mechanisms of Hb degradation and Hz formation are poorly understood. We report the presence of a ∼200-kDa protein complex in the food vacuole that is required for Hb degradation and Hz formation. This complex contains several parasite proteins, including falcipain 2/2', plasmepsin II, plasmepsin IV, histo aspartic protease, and heme detoxification protein. The association of these proteins is evident from coimmunoprecipitation followed by mass spectrometry, coelution from a gel filtration column, cosedimentation on a glycerol gradient, and in vitro protein interaction analyses. To functionally characterize this complex, we developed an in vitro assay using two of the proteins present in the complex. Our results show that falcipain 2 and heme detoxification protein associate with each other to efficiently convert Hb to Hz. We also used this in vitro assay to elucidate the modes of action of chloroquine and artemisinin. Our results reveal that both chloroquine and artemisinin act during the heme polymerization step, and chloroquine also acts at the Hb degradation step. These results may have important implications in the development of previously undefined antimalarials.


Asunto(s)
Antimaláricos/farmacología , Cisteína Endopeptidasas/metabolismo , Hemoproteínas/biosíntesis , Hemoglobinas/metabolismo , Complejos Multiproteicos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Artemisininas , Cloroquina , Cromatografía en Gel , Inmunoprecipitación , Espectrometría de Masas , Polimerizacion/efectos de los fármacos , Proteolisis/efectos de los fármacos
17.
Eukaryot Cell ; 13(4): 504-16, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24562907

RESUMEN

Human African trypanosomiasis (HAT) is an important public health threat in sub-Saharan Africa. Current drugs are unsatisfactory, and new drugs are being sought. Few validated enzyme targets are available to support drug discovery efforts, so our goal was to obtain essentiality data on genes with proven utility as drug targets. Aminoacyl-tRNA synthetases (aaRSs) are known drug targets for bacterial and fungal pathogens and are required for protein synthesis. Here we survey the essentiality of eight Trypanosoma brucei aaRSs by RNA interference (RNAi) gene expression knockdown, covering an enzyme from each major aaRS class: valyl-tRNA synthetase (ValRS) (class Ia), tryptophanyl-tRNA synthetase (TrpRS-1) (class Ib), arginyl-tRNA synthetase (ArgRS) (class Ic), glutamyl-tRNA synthetase (GluRS) (class 1c), threonyl-tRNA synthetase (ThrRS) (class IIa), asparaginyl-tRNA synthetase (AsnRS) (class IIb), and phenylalanyl-tRNA synthetase (α and ß) (PheRS) (class IIc). Knockdown of mRNA encoding these enzymes in T. brucei mammalian stage parasites showed that all were essential for parasite growth and survival in vitro. The reduced expression resulted in growth, morphological, cell cycle, and DNA content abnormalities. ThrRS was characterized in greater detail, showing that the purified recombinant enzyme displayed ThrRS activity and that the protein localized to both the cytosol and mitochondrion. Borrelidin, a known inhibitor of ThrRS, was an inhibitor of T. brucei ThrRS and showed antitrypanosomal activity. The data show that aaRSs are essential for T. brucei survival and are likely to be excellent targets for drug discovery efforts.


Asunto(s)
Aminoacil-ARNt Sintetasas/antagonistas & inhibidores , Proteínas Protozoarias/antagonistas & inhibidores , ARN Mensajero/antagonistas & inhibidores , Tripanocidas/farmacología , Trypanosoma brucei brucei/efectos de los fármacos , Aminoacil-ARNt Sintetasas/genética , Aminoacil-ARNt Sintetasas/metabolismo , Ciclo Celular/efectos de los fármacos , Citosol/efectos de los fármacos , Citosol/enzimología , Descubrimiento de Drogas , Alcoholes Grasos/farmacología , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Trypanosoma brucei brucei/enzimología , Trypanosoma brucei brucei/genética
18.
J Biol Chem ; 288(20): 14256-14263, 2013 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-23548908

RESUMEN

Trypanosoma brucei sp. causes human African trypanosomiasis (HAT; African sleeping sickness). The parasites initially proliferate in the hemolymphatic system and then invade the central nervous system, which is lethal if not treated. New drugs are needed for HAT because the approved drugs are few, toxic, and difficult to administer, and drug resistance is spreading. We showed by RNAi knockdown that T. brucei isoleucyl-tRNA synthetase is essential for the parasites in vitro and in vivo in a mouse model of infection. By structure prediction and experimental analysis, we also identified small molecules that inhibit recombinant isoleucyl-tRNA synthetase and that are lethal to the parasites in vitro and highly selective compared with mammalian cells. One of these molecules acts as a competitive inhibitor of the enzyme and cures mice of the infection. Because members of this class of molecules are known to cross the blood-brain barrier in humans and to be tolerated, they may be attractive as leading candidates for drug development for HAT.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Isoleucina-ARNt Ligasa/antagonistas & inhibidores , Tripanocidas/farmacología , Trypanosoma brucei brucei/enzimología , Tripanosomiasis Africana/tratamiento farmacológico , Animales , Barrera Hematoencefálica , Modelos Animales de Enfermedad , Diseño de Fármacos , Inhibidores Enzimáticos/uso terapéutico , Isoleucina-ARNt Ligasa/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Interferencia de ARN , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Transfección , Tripanocidas/uso terapéutico
19.
RNA ; 18(2): 308-20, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22184461

RESUMEN

Three distinct editosomes are required for the uridine insertion/deletion editing that creates translatable mitochondrial mRNAs in Trypanosoma brucei. They contain KREPB6, KREPB7, or KREPB8 proteins and their respective endonucleases KREN3, KREN2, or KREN1. RNAi knockdowns of KREPB6, KREPB7, and KREPB8 variably affect growth and RNA editing. KREPB6 and KREPB7 knockdowns substantially reduced in vitro insertion site cleavage activity of their respective editosomes, while KREPB8 knockdown did not affect its editosome deletion site cleavage activity despite inhibition of growth and editing. KREPB6, KREPB7, and KREPB8 knockdowns disrupted tagged KREN3, KREN2, or KREN1 editosomes, respectively, to varying degrees, and in the case of KREN1 editosomes, the deletion editing site cleavage activity shifted to a smaller S value. The varying effects correlate with a combination of the relative abundances of the KREPB6-8 proteins and of the different insertion and deletion sites. Tagged KREPB6-8 were physically associated with deletion subcomplexes upon knockdown of the centrally interactive KREPA3 protein, while KREN1-3 endonucleases were associated with insertion subcomplexes. The results indicate that KREPB6-8 occupy similar positions in editosomes and are important for the activity and specificity of their respective endonucleases. This suggests that they contribute to the accurate recognition of the numerous similar but diverse editing site substrates.


Asunto(s)
Endonucleasas/genética , Endonucleasas/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Edición de ARN , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/metabolismo , Técnicas de Silenciamiento del Gen/métodos , Mutagénesis Insercional , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mitocondrial , ARN Protozoario/genética , Trypanosoma brucei brucei/enzimología , Trypanosoma brucei brucei/crecimiento & desarrollo , Uridina/metabolismo
20.
RNA ; 18(10): 1897-909, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22919050

RESUMEN

The transcriptome of kinetoplastid mitochondria undergoes extensive RNA editing that inserts and deletes uridine residues (U's) to produce mature mRNAs. The editosome is a multiprotein complex that provides endonuclease, TUTase, exonuclease, and ligase activities required for RNA editing. The editosome's KREPB4 and KREPB5 proteins are essential for editosome integrity and parasite viability and contain semi-conserved motifs corresponding to zinc finger, RNase III, and PUF domains, but to date no functional analysis of these domains has been reported. We show here that various point mutations to KREPB4 and KREPB5 identify essential domains, and suggest that these proteins do not themselves perform RNase III catalysis. The zinc finger of KREPB4 but not KREPB5 is essential for editosome integrity and parasite viability, and mutation of the RNase III signature motif in KREPB5 prevents integration into editosomes, which is lethal. Isolated TAP-tagged KREPB4 and KREPB5 complexes preferentially associate with components of the deletion subcomplex, providing additional insights into editosome architecture. A new alignment of editosome RNase III sequences from several kinetoplastid species implies that KREPB4 and KREPB5 lack catalytic activity and reveals that the PUF motif is present in the editing endonucleases KREN1, KREN2, and KREN3. The data presented here are consistent with the hypothesis that KREPB4 and KREPB5 form intermolecular heterodimers with the catalytically active editing endonucleases, which is unprecedented among known RNase III proteins.


Asunto(s)
Edición de ARN/genética , ARN Protozoario/genética , Ribonucleoproteínas/genética , Ribonucleoproteínas/fisiología , Trypanosoma brucei brucei/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos/fisiología , Dominio Catalítico/genética , Análisis Mutacional de ADN , Genoma de Protozoos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , ARN Mensajero/análisis , ARN Mensajero/genética , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo , Homología de Secuencia , Trypanosoma brucei brucei/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda