Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Nat Immunol ; 20(5): 571-580, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30936493

RESUMEN

Fine control of macrophage activation is needed to prevent inflammatory disease, particularly at barrier sites such as the lungs. However, the dominant mechanisms that regulate the activation of pulmonary macrophages during inflammation are poorly understood. We found that alveolar macrophages (AlvMs) were much less able to respond to the canonical type 2 cytokine IL-4, which underpins allergic disease and parasitic worm infections, than macrophages from lung tissue or the peritoneal cavity. We found that the hyporesponsiveness of AlvMs to IL-4 depended upon the lung environment but was independent of the host microbiota or the lung extracellular matrix components surfactant protein D (SP-D) and mucin 5b (Muc5b). AlvMs showed severely dysregulated metabolism relative to that of cavity macrophages. After removal from the lungs, AlvMs regained responsiveness to IL-4 in a glycolysis-dependent manner. Thus, impaired glycolysis in the pulmonary niche regulates AlvM responsiveness during type 2 inflammation.


Asunto(s)
Inflamación/inmunología , Pulmón/inmunología , Activación de Macrófagos/inmunología , Macrófagos Alveolares/inmunología , Animales , Inflamación/genética , Inflamación/metabolismo , Interleucina-4/genética , Interleucina-4/inmunología , Interleucina-4/metabolismo , Larva/inmunología , Larva/fisiología , Pulmón/metabolismo , Pulmón/patología , Activación de Macrófagos/genética , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/parasitología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mucina 5B/genética , Mucina 5B/inmunología , Mucina 5B/metabolismo , Nippostrongylus/inmunología , Nippostrongylus/fisiología , Proteína D Asociada a Surfactante Pulmonar/genética , Proteína D Asociada a Surfactante Pulmonar/inmunología , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Infecciones por Strongylida/genética , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología
2.
PLoS Pathog ; 20(2): e1011889, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38408115

RESUMEN

Trypanosomatid parasites undergo developmental regulation to adapt to the different environments encountered during their life cycle. In Trypanosoma brucei, a genome wide selectional screen previously identified a regulator of the protein family ESAG9, which is highly expressed in stumpy forms, a morphologically distinct bloodstream stage adapted for tsetse transmission. This regulator, TbREG9.1, has an orthologue in Trypanosoma congolense, despite the absence of a stumpy morphotype in that parasite species, which is an important cause of livestock trypanosomosis. RNAi mediated gene silencing of TcREG9.1 in Trypanosoma congolense caused a loss of attachment of the parasites to a surface substrate in vitro, a key feature of the biology of these parasites that is distinct from T. brucei. This detachment was phenocopied by treatment of the parasites with a phosphodiesterase inhibitor, which also promotes detachment in the insect trypanosomatid Crithidia fasciculata. RNAseq analysis revealed that TcREG9.1 silencing caused the upregulation of mRNAs for several classes of surface molecules, including transferrin receptor-like molecules, immunoreactive proteins in experimental bovine infections, and molecules related to those associated with stumpy development in T. brucei. Depletion of TcREG9.1 in vivo also generated an enhanced level of parasites in the blood circulation consistent with reduced parasite attachment to the microvasculature. The morphological progression to insect forms of the parasite was also perturbed. We propose a model whereby TcREG9.1 acts as a regulator of attachment and development, with detached parasites being adapted for transmission.


Asunto(s)
Trypanosoma brucei brucei , Trypanosoma congolense , Animales , Bovinos , Trypanosoma brucei brucei/fisiología , Interferencia de ARN , Silenciador del Gen
3.
Immunity ; 47(4): 739-751.e5, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-29045903

RESUMEN

Infection by helminth parasites is associated with amelioration of allergic reactivity, but mechanistic insights into this association are lacking. Products secreted by the mouse parasite Heligmosomoides polygyrus suppress type 2 (allergic) immune responses through interference in the interleukin-33 (IL-33) pathway. Here, we identified H. polygyrus Alarmin Release Inhibitor (HpARI), an IL-33-suppressive 26-kDa protein, containing three predicted complement control protein (CCP) modules. In vivo, recombinant HpARI abrogated IL-33, group 2 innate lymphoid cell (ILC2) and eosinophilic responses to Alternaria allergen administration, and diminished eosinophilic responses to Nippostrongylus brasiliensis, increasing parasite burden. HpARI bound directly to both mouse and human IL-33 (in the cytokine's activated state) and also to nuclear DNA via its N-terminal CCP module pair (CCP1/2), tethering active IL-33 within necrotic cells, preventing its release, and forestalling initiation of type 2 allergic responses. Thus, HpARI employs a novel molecular strategy to suppress type 2 immunity in both infection and allergy.


Asunto(s)
Proteínas del Helminto/inmunología , Interleucina-33/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Alérgenos/inmunología , Alternaria/inmunología , Secuencia de Aminoácidos , Animales , Western Blotting , Eosinófilos/inmunología , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Interacciones Huésped-Parásitos/inmunología , Humanos , Inmunidad Innata/inmunología , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33/genética , Interleucina-33/metabolismo , Linfocitos/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Nematospiroides dubius/genética , Nematospiroides dubius/metabolismo , Unión Proteica/inmunología , Receptores de Interleucina/inmunología , Receptores de Interleucina/metabolismo , Homología de Secuencia de Aminoácido , Infecciones por Strongylida/metabolismo , Infecciones por Strongylida/parasitología
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.
RNA ; 26(12): 1862-1881, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32873716

RESUMEN

Trypanosome U-insertion/deletion RNA editing in mitochondrial mRNAs involves guide RNAs (gRNAs) and the auxiliary RNA editing substrate binding complex (RESC) and RNA editing helicase 2 complex (REH2C). RESC and REH2C stably copurify with editing mRNAs but the functional interplay between these complexes remains unclear. Most steady-state mRNAs are partially edited and include misedited "junction" regions that match neither pre-mRNA nor fully edited transcripts. Editing specificity is central to mitochondrial RNA maturation and function, but its basic control mechanisms remain unclear. Here we applied a novel nucleotide-resolution RNA-seq approach to examine ribosomal protein subunit 12 (RPS12) and ATPase subunit 6 (A6) mRNA transcripts. We directly compared transcripts associated with RESC and REH2C to those found in total mitochondrial RNA. RESC-associated transcripts exhibited site-preferential enrichments in total and accurate edits. REH2C loss-of-function induced similar substrate-specific and site-specific editing effects in total and RESC-associated RNA. It decreased total editing primarily at RPS12 5' positions but increased total editing at examined A6 3' positions. REH2C loss-of-function caused site-preferential loss of accurate editing in both transcripts. However, changes in total or accurate edits did not necessarily involve common sites. A few 5' nucleotides of the initiating gRNA (gRNA-1) directed accurate editing in both transcripts. However, in RPS12, two conserved 3'-terminal adenines in gRNA-1 could direct a noncanonical 2U-insertion that causes major pausing in 3'-5' progression. In A6, a noncanonical sequence element that depends on REH2C in a region normally targeted by the 3' half of gRNA-1 may hinder early editing progression. Overall, we defined transcript-specific effects of REH2C loss.


Asunto(s)
Proteínas Protozoarias/metabolismo , Edición de ARN , ARN Mensajero/metabolismo , ARN Mitocondrial/metabolismo , ARN Protozoario/metabolismo , Trypanosoma brucei brucei/metabolismo , Trypanosoma/metabolismo , Animales , Proteínas Protozoarias/genética , ARN Guía de Kinetoplastida , ARN Mensajero/genética , ARN Mitocondrial/genética , ARN Protozoario/genética , RNA-Seq , Especificidad por Sustrato , Trypanosoma/genética , Trypanosoma brucei brucei/genética
6.
Proc Natl Acad Sci U S A ; 116(45): 22774-22782, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31636179

RESUMEN

African trypanosomes use an extreme form of antigenic variation to evade host immunity, involving the switching of expressed variant surface glycoproteins by a stochastic and parasite-intrinsic process. Parasite development in the mammalian host is another feature of the infection dynamic, with trypanosomes undergoing quorum sensing (QS)-dependent differentiation between proliferative slender forms and arrested, transmissible, stumpy forms. Longstanding experimental studies have suggested that the frequency of antigenic variation and transmissibility may be linked, antigen switching being higher in developmentally competent, fly-transmissible, parasites ("pleomorphs") than in serially passaged "monomorphic" lines that cannot transmit through flies. Here, we have directly tested this tenet of the infection dynamic by using 2 experimental systems to reduce pleomorphism. Firstly, lines were generated that inducibly lose developmental capacity through RNAi-mediated silencing of the QS signaling machinery ("inducible monomorphs"). Secondly, de novo lines were derived that have lost the capacity for stumpy formation by serial passage ("selected monomorphs") and analyzed for their antigenic variation in comparison to isogenic preselected populations. Analysis of both inducible and selected monomorphs has established that antigen switch frequency and developmental capacity are independently selected traits. This generates the potential for diverse infection dynamics in different parasite populations where the rate of antigenic switching and transmission competence are uncoupled. Further, this may support the evolution, maintenance, and spread of important trypanosome variants such as Trypanosoma brucei evansi that exploit mechanical transmission.


Asunto(s)
Antígenos de Protozoos/inmunología , Trypanosoma brucei brucei/inmunología , Animales , Antígenos de Protozoos/genética , Interferencia de ARN , Glicoproteínas Variantes de Superficie de Trypanosoma/genética
7.
Proc Natl Acad Sci U S A ; 116(39): 19753-19759, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31506353

RESUMEN

Hypoxia is a ubiquitous feature of cancers, encouraging glycolytic metabolism, proliferation, and resistance to therapy. Nonetheless, hypoxia is a poorly defined term with confounding features described in the literature. Redox biology provides an important link between the external cellular microenvironment and the cell's response to changing oxygen pressures. In this paper, we demonstrate a correlation between intracellular redox potential (measured using optical nanosensors) and the concentrations of microRNAs (miRNAs) involved in the cell's response to changes in oxygen pressure. The correlations were established using surprisal analysis (an approach derived from thermodynamics and information theory). We found that measured redox potential changes reflect changes in the free energy computed by surprisal analysis of miRNAs. Furthermore, surprisal analysis identified groups of miRNAs, functionally related to changes in proliferation and metastatic potential that played the most significant role in the cell's response to changing oxygen pressure.


Asunto(s)
Hipoxia de la Célula/genética , Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/genética , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Citoplasma/metabolismo , Humanos , Hipoxia/metabolismo , Células MCF-7/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno , Termodinámica , Microambiente Tumoral/genética
8.
J Virol ; 94(21)2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-32796064

RESUMEN

African swine fever virus (ASFV) causes a lethal hemorrhagic disease of domestic pigs, against which no vaccine is available. ASFV has a large, double-stranded DNA genome that encodes over 150 proteins. Replication takes place predominantly in the cytoplasm of the cell and involves complex interactions with host cellular components, including small noncoding RNAs (sncRNAs). A number of DNA viruses are known to manipulate sncRNA either by encoding their own or disrupting host sncRNA. To investigate the interplay between ASFV and sncRNAs, a study of host and viral small RNAs extracted from ASFV-infected primary porcine macrophages (PAMs) was undertaken. We discovered that ASFV infection had only a modest effect on host miRNAs, with only 6 miRNAs differentially expressed during infection. The data also revealed 3 potential novel small RNAs encoded by ASFV, ASFVsRNA1-3. Further investigation of ASFVsRNA2 detected it in lymphoid tissue from pigs with ASF. Overexpression of ASFVsRNA2 led to an up to 1-log reduction in ASFV growth, indicating that ASFV utilizes a virus-encoded small RNA to disrupt its own replication.IMPORTANCE African swine fever (ASF) poses a major threat to pig populations and food security worldwide. The disease is endemic to Africa and Eastern Europe and is rapidly emerging into Asia, where it has led to the deaths of millions of pigs in the last 12 months. The development of safe and effective vaccines to protect pigs against ASF has been hindered by lack of understanding of the complex interactions between ASFV and the host cell. We focused our work on characterizing the interactions between ASFV and sncRNAs. Although comparatively modest changes to host sncRNA abundances were observed upon ASFV infection, we discovered and characterized a novel functional ASFV-encoded sncRNA. The results from this study add important insights into ASFV host-pathogen interactions. This knowledge may be exploited to develop more effective ASFV vaccines that take advantage of the sncRNA system.


Asunto(s)
Virus de la Fiebre Porcina Africana/genética , Fiebre Porcina Africana/genética , Genoma Viral , Interacciones Huésped-Patógeno/genética , MicroARNs/genética , ARN Pequeño no Traducido/genética , ARN Viral/genética , Fiebre Porcina Africana/metabolismo , Fiebre Porcina Africana/virología , Virus de la Fiebre Porcina Africana/metabolismo , Animales , Regulación de la Expresión Génica , Tamaño del Genoma , Tejido Linfoide , Macrófagos , MicroARNs/clasificación , MicroARNs/metabolismo , Cultivo Primario de Células , ARN Pequeño no Traducido/clasificación , ARN Pequeño no Traducido/metabolismo , ARN Viral/clasificación , ARN Viral/metabolismo , Transducción de Señal , Sus scrofa , Porcinos , Replicación Viral
9.
Nucleic Acids Res ; 47(21): 11304-11325, 2019 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-31665448

RESUMEN

Kinetoplastids are protists defined by one of the most complex mitochondrial genomes in nature, the kinetoplast. In the sleeping sickness parasite Trypanosoma brucei, the kinetoplast is a chain mail-like network of two types of interlocked DNA molecules: a few dozen ∼23-kb maxicircles (homologs of the mitochondrial genome of other eukaryotes) and thousands of ∼1-kb minicircles. Maxicircles encode components of respiratory chain complexes and the mitoribosome. Several maxicircle-encoded mRNAs undergo extensive post-transcriptional RNA editing via addition and deletion of uridines. The process is mediated by hundreds of species of minicircle-encoded guide RNAs (gRNAs), but the precise number of minicircle classes and gRNA genes was unknown. Here we present the first essentially complete assembly and annotation of the kinetoplast genome of T. brucei. We have identified 391 minicircles, encoding not only ∼930 predicted 'canonical' gRNA genes that cover nearly all known editing events (accessible via the web at http://hank.bio.ed.ac.uk), but also ∼370 'non-canonical' gRNA genes of unknown function. Small RNA transcriptome data confirmed expression of the majority of both categories of gRNAs. Finally, we have used our data set to refine definitions for minicircle structure and to explore dynamics of minicircle copy numbers.


Asunto(s)
Genoma Mitocondrial , Anotación de Secuencia Molecular , Análisis de Secuencia de ADN , Trypanosoma brucei brucei/genética , Animales , Secuencia de Bases , Secuencia Conservada , ADN Circular/análisis , ADN Circular/genética , ADN de Cinetoplasto/genética , Orden Génico , Genoma de Protozoos , ARN Guía de Kinetoplastida/genética , Trypanosoma brucei brucei/ultraestructura
10.
PLoS Pathog ; 14(6): e1007145, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29940034

RESUMEN

Trypanosoma brucei, the agents of African trypanosomiasis, undergo density-dependent differentiation in the mammalian bloodstream to prepare for transmission by tsetse flies. This involves the generation of cell-cycle arrested, quiescent, stumpy forms from proliferative slender forms. The signalling pathway responsible for the quorum sensing response has been catalogued using a genome-wide selective screen, providing a compendium of signalling protein kinases phosphatases, RNA binding proteins and hypothetical proteins. However, the ordering of these components is unknown. To piece together these components to provide a description of how stumpy formation arises we have used an extragenic suppression approach. This exploited a combinatorial gene knockout and overexpression strategy to assess whether the loss of developmental competence in null mutants of pathway components could be compensated by ectopic expression of other components. We have created null mutants for three genes in the stumpy induction factor signalling pathway (RBP7, YAK, MEKK1) and evaluated complementation by expression of RBP7, NEK17, PP1-6, or inducible gene silencing of the proposed differentiation inhibitor TbTOR4. This indicated that the signalling pathway is non-linear. Phosphoproteomic analysis focused on one pathway component, a putative MEKK, identified molecules with altered expression and phosphorylation profiles in MEKK1 null mutants, including another component in the pathway, NEK17. Our data provide a first molecular dissection of multiple components in a signal transduction cascade in trypanosomes.


Asunto(s)
Sangre/parasitología , Proteínas Protozoarias/metabolismo , Percepción de Quorum , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Trypanosoma brucei brucei/metabolismo , Tripanosomiasis Africana/parasitología , Animales , Diferenciación Celular , Genoma , Ratones , Fosforilación , Proteínas Protozoarias/genética , Proteínas de Unión al ARN/genética , Trypanosoma brucei brucei/genética
11.
Nature ; 505(7485): 681-685, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24336212

RESUMEN

The protozoan parasites Trypanosoma brucei spp. cause important human and livestock diseases in sub-Saharan Africa. In mammalian blood, two developmental forms of the parasite exist: proliferative 'slender' forms and arrested 'stumpy' forms that are responsible for transmission to tsetse flies. The slender to stumpy differentiation is a density-dependent response that resembles quorum sensing in microbial systems and is crucial for the parasite life cycle, ensuring both infection chronicity and disease transmission. This response is triggered by an elusive 'stumpy induction factor' (SIF) whose intracellular signalling pathway is also uncharacterized. Laboratory-adapted (monomorphic) trypanosome strains respond inefficiently to SIF but can generate forms with stumpy characteristics when exposed to cell-permeable cAMP and AMP analogues. Exploiting this, we have used a genome-wide RNA interference library screen to identify the signalling components driving stumpy formation. In separate screens, monomorphic parasites were exposed to 8-(4-chlorophenylthio)-cAMP (pCPT-cAMP) or 8-pCPT-2'-O-methyl-5'-AMP to select cells that were unresponsive to these signals and hence remained proliferative. Genome-wide Ion Torrent based RNAi target sequencing identified cohorts of genes implicated in each step of the signalling pathway, from purine metabolism, through signal transducers (kinases, phosphatases) to gene expression regulators. Genes at each step were independently validated in cells naturally capable of stumpy formation, confirming their role in density sensing in vivo. The putative RNA-binding protein, RBP7, was required for normal quorum sensing and promoted cell-cycle arrest and transmission competence when overexpressed. This study reveals that quorum sensing signalling in trypanosomes shares similarities to fundamental quiescence pathways in eukaryotic cells, its components providing targets for quorum-sensing interference-based therapeutics.


Asunto(s)
Genoma/genética , Percepción de Quorum/genética , Transducción de Señal/genética , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/metabolismo , Animales , Diferenciación Celular , AMP Cíclico/metabolismo , Fase G1 , Puntos de Control de la Fase G1 del Ciclo Celular , Regulación de la Expresión Génica , Proteínas Quinasas/genética , Interferencia de ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Reproducibilidad de los Resultados , Trypanosoma brucei brucei/enzimología , Trypanosoma brucei brucei/crecimiento & desarrollo
12.
PLoS Pathog ; 13(3): e1006279, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28334017

RESUMEN

Trypanosoma brucei, causing African sleeping-sickness, exploits quorum-sensing (QS) to generate the 'stumpy forms' necessary for the parasite's transmission to tsetse-flies. These quiescent cells are generated by differentiation in the bloodstream from proliferative slender forms. Using genome-wide RNAi selection we screened for repressors of transmission stage-enriched mRNAs in slender forms, using the stumpy-elevated ESAG9 transcript as a model. This identified REG9.1, whose RNAi-silencing alleviated ESAG9 repression in slender forms and tsetse-midgut procyclic forms. Interestingly, trypanosome surface protein Family 5 and Family 7 mRNAs were also elevated, which, like ESAG9, are T. brucei specific and stumpy-enriched. We suggest these contribute to the distinct transmission biology and vector tropism of T. brucei from other African trypanosome species. As well as surface family regulation, REG9.1-depletion generated QS-independent development to stumpy forms in vivo, whereas REG9.1 overexpression in bloodstream forms potentiated spontaneous differentiation to procyclic forms in the absence of an external signal. Combined, this identifies REG9.1 as a regulator of developmental cell fate, controlling the expression of Trypanosoma brucei-specific molecules elevated during transmission.


Asunto(s)
Regulación de la Expresión Génica/genética , Proteínas Protozoarias/biosíntesis , Proteínas de Unión al ARN/biosíntesis , Tripanosomiasis Africana/genética , Tripanosomiasis Africana/transmisión , Animales , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Estudio de Asociación del Genoma Completo , Immunoblotting , Ratones , Proteínas Protozoarias/genética , Interferencia de ARN , Proteínas de Unión al ARN/genética , Transfección , Trypanosoma brucei brucei , Trypanosoma congolense , Trypanosoma vivax
13.
Genome Res ; 24(10): 1676-85, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25015382

RESUMEN

Global production of chickens has trebled in the past two decades and they are now the most important source of dietary animal protein worldwide. Chickens are subject to many infectious diseases that reduce their performance and productivity. Coccidiosis, caused by apicomplexan protozoa of the genus Eimeria, is one of the most important poultry diseases. Understanding the biology of Eimeria parasites underpins development of new drugs and vaccines needed to improve global food security. We have produced annotated genome sequences of all seven species of Eimeria that infect domestic chickens, which reveal the full extent of previously described repeat-rich and repeat-poor regions and show that these parasites possess the most repeat-rich proteomes ever described. Furthermore, while no other apicomplexan has been found to possess retrotransposons, Eimeria is home to a family of chromoviruses. Analysis of Eimeria genes involved in basic biology and host-parasite interaction highlights adaptations to a relatively simple developmental life cycle and a complex array of co-expressed surface proteins involved in host cell binding.


Asunto(s)
Eimeria/genética , Genoma de Protozoos , Proteínas Protozoarias/genética , Animales , Línea Celular , Pollos , Mapeo Cromosómico , Coccidiosis/parasitología , Coccidiosis/veterinaria , Eimeria/clasificación , Perfilación de la Expresión Génica , Filogenia , Enfermedades de las Aves de Corral/parasitología , Proteoma , Sintenía
14.
Nucleic Acids Res ; 43(9): 4491-504, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25873624

RESUMEN

Trypanosomes mostly regulate gene expression through post-transcriptional mechanisms, particularly mRNA stability. However, much mRNA degradation is cytoplasmic such that mRNA nuclear export must represent an important level of regulation. Ribosomal RNAs must also be exported from the nucleus and the trypanosome orthologue of NMD3 has been confirmed to be involved in rRNA processing and export, matching its function in other organisms. Surprisingly, we found that TbNMD3 depletion also generates mRNA accumulation of procyclin-associated genes (PAGs), these being co-transcribed by RNA polymerase I with the procyclin surface antigen genes expressed on trypanosome insect forms. By whole transcriptome RNA-seq analysis of TbNMD3-depleted cells we confirm the regulation of the PAG transcripts by TbNMD3 and using reporter constructs reveal that PAG1 regulation is mediated by its 5'UTR. Dissection of the mechanism of regulation demonstrates that it is not dependent upon translational inhibition mediated by TbNMD3 depletion nor enhanced transcription. However, depletion of the nuclear export factors XPO1 or MEX67 recapitulates the effects of TbNMD3 depletion on PAG mRNAs and mRNAs accumulated in the nucleus of TbNMD3-depleted cells. These results invoke a novel RNA regulatory mechanism involving the NMD3-dependent nuclear export of mRNA cargos, suggesting a shared platform for mRNA and rRNA export.


Asunto(s)
Núcleo Celular/metabolismo , Carioferinas/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Proteínas Protozoarias/fisiología , ARN Mensajero/metabolismo , ARN Ribosómico/metabolismo , Proteínas de Unión al ARN/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Regiones no Traducidas 5' , Transporte Activo de Núcleo Celular , Carioferinas/antagonistas & inhibidores , Proteínas de Transporte Nucleocitoplasmático/antagonistas & inhibidores , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas de Unión al ARN/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Activación Transcripcional , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/metabolismo , Proteína Exportina 1
15.
Proc Natl Acad Sci U S A ; 111(11): 4280-4, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24594597

RESUMEN

Quorum sensing (QS) is a cell-cell communication system that controls gene expression in many bacterial species, mediated by diffusible signal molecules. Although the intracellular regulatory mechanisms of QS are often well-understood, the functional roles of QS remain controversial. In particular, the use of multiple signals by many bacterial species poses a serious challenge to current functional theories. Here, we address this challenge by showing that bacteria can use multiple QS signals to infer both their social (density) and physical (mass-transfer) environment. Analytical and evolutionary simulation models show that the detection of, and response to, complex social/physical contrasts requires multiple signals with distinct half-lives and combinatorial (nonadditive) responses to signal concentrations. We test these predictions using the opportunistic pathogen Pseudomonas aeruginosa and demonstrate significant differences in signal decay between its two primary signal molecules, as well as diverse combinatorial responses to dual-signal inputs. QS is associated with the control of secreted factors, and we show that secretome genes are preferentially controlled by synergistic "AND-gate" responses to multiple signal inputs, ensuring the effective expression of secreted factors in high-density and low mass-transfer environments. Our results support a new functional hypothesis for the use of multiple signals and, more generally, show that bacteria are capable of combinatorial communication.


Asunto(s)
Fenómenos Fisiológicos Bacterianos , Ambiente , Regulación Bacteriana de la Expresión Génica/fisiología , Modelos Biológicos , Percepción de Quorum/fisiología , Biología Computacional , Simulación por Computador , Análisis por Micromatrices , Densidad de Población , Pseudomonas aeruginosa
16.
Int Immunol ; 27(11): 589-96, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25899567

RESUMEN

The archetypal Th2 cytokine IL-4 has previously been shown to alternatively activate murine macrophages and, more recently, dendritic cells (DCs) both in vitro and in vivo. IL-4 has also been shown to induce Aldh1a2 (aldehyde dehydrogenase 1a2) expression in murine macrophages recruited to the peritoneal cavity. However, the influence of IL-4 on DC Aldh1a2 induction in vivo has not yet been addressed. In this work, we found that DCs show enhanced aldehyde dehydrogenase enzyme activity in vivo, which led us to investigate the impact of the vitamin A metabolite all-trans retinoic acid (RA) on DC alternative activation and function. Antagonism of RA receptors reduced production of resistin-like molecule alpha by DCs responding to IL-4, while addition of exogenous RA enhanced production of this marker of alternative activation. Functionally, RA increased DC induction of CD4(+) T-cell IL-10, while reducing CD4(+) T-cell IL-4 and IL-13, revealing a previously unidentified role for RA in regulating the ability of alternatively activated DCs to influence Th2 polarization.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Inmunomodulación/efectos de los fármacos , Tretinoina/farmacología , Aldehído Deshidrogenasa/metabolismo , Animales , Antígenos de Superficie/metabolismo , Células Dendríticas/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Inmunofenotipificación , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-4/farmacología , Ratones , Fenotipo , Receptores de Ácido Retinoico/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo
17.
Nat Genet ; 39(7): 839-47, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17572675

RESUMEN

Leishmania parasites cause a broad spectrum of clinical disease. Here we report the sequencing of the genomes of two species of Leishmania: Leishmania infantum and Leishmania braziliensis. The comparison of these sequences with the published genome of Leishmania major reveals marked conservation of synteny and identifies only approximately 200 genes with a differential distribution between the three species. L. braziliensis, contrary to Leishmania species examined so far, possesses components of a putative RNA-mediated interference pathway, telomere-associated transposable elements and spliced leader-associated SLACS retrotransposons. We show that pseudogene formation and gene loss are the principal forces shaping the different genomes. Genes that are differentially distributed between the species encode proteins implicated in host-pathogen interactions and parasite survival in the macrophage.


Asunto(s)
Genoma , Genómica , Leishmania/genética , Leishmaniasis/parasitología , Secuencia de Aminoácidos , Animales , Humanos , Leishmania braziliensis/genética , Leishmania infantum/genética , Leishmania major/genética , Leishmaniasis Cutánea/parasitología , Leishmaniasis Visceral/parasitología , Datos de Secuencia Molecular
18.
Antimicrob Agents Chemother ; 59(6): 3098-108, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25779578

RESUMEN

Overexpression of the ABC transporter genes patA and patB confers efflux-mediated fluoroquinolone resistance in Streptococcus pneumoniae and is also linked to pneumococcal stress responses. Although upregulation of patAB has been observed in many laboratory mutants and clinical isolates, the regulatory mechanisms controlling expression of these genes are unknown. In this study, we aimed to identify the cause of high-level constitutive overexpression of patAB in M184, a multidrug-resistant mutant of S. pneumoniae R6. Using a whole-genome transformation and sequencing approach, we identified a novel duplication of a 9.2-kb region of the M184 genome which included the patAB genes. This duplication did not affect growth and was semistable with a low segregation rate. The expression levels of patAB in M184 were much higher than those that could be fully explained by doubling of the gene dosage alone, and inactivation of the first copy of patA had no effect on multidrug resistance. Using a green fluorescent protein reporter system, increased patAB expression was ascribed to transcriptional read-through from a tRNA gene upstream of the second copy of patAB. This is the first report of a large genomic duplication causing antibiotic resistance in S. pneumoniae and also of a genomic duplication causing antibiotic resistance by a promoter switching mechanism.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Proteínas Bacterianas/genética , Fluoroquinolonas/farmacología , Amplificación de Genes/genética , Streptococcus pneumoniae/genética , Antibacterianos/farmacología , Streptococcus pneumoniae/efectos de los fármacos
19.
Nature ; 460(7253): 352-8, 2009 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-19606141

RESUMEN

Schistosoma mansoni is responsible for the neglected tropical disease schistosomiasis that affects 210 million people in 76 countries. Here we present analysis of the 363 megabase nuclear genome of the blood fluke. It encodes at least 11,809 genes, with an unusual intron size distribution, and new families of micro-exon genes that undergo frequent alternative splicing. As the first sequenced flatworm, and a representative of the Lophotrochozoa, it offers insights into early events in the evolution of the animals, including the development of a body pattern with bilateral symmetry, and the development of tissues into organs. Our analysis has been informed by the need to find new drug targets. The deficits in lipid metabolism that make schistosomes dependent on the host are revealed, and the identification of membrane receptors, ion channels and more than 300 proteases provide new insights into the biology of the life cycle and new targets. Bioinformatics approaches have identified metabolic chokepoints, and a chemogenomic screen has pinpointed schistosome proteins for which existing drugs may be active. The information generated provides an invaluable resource for the research community to develop much needed new control tools for the treatment and eradication of this important and neglected disease.


Asunto(s)
Genoma de los Helmintos/genética , Schistosoma mansoni/genética , Animales , Evolución Biológica , Exones/genética , Genes de Helminto/genética , Interacciones Huésped-Parásitos/genética , Intrones/genética , Datos de Secuencia Molecular , Mapeo Físico de Cromosoma , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/embriología , Schistosoma mansoni/fisiología , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/parasitología
20.
Eukaryot Cell ; 13(3): 412-26, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24442893

RESUMEN

In the bloodstream of mammalian hosts, the sleeping sickness parasite, Trypanosoma brucei, exists as a proliferative slender form or a nonproliferative, transmissible, stumpy form. The transition between these developmental forms is controlled by a density-dependent mechanism that is important for the parasite's infection dynamics, immune evasion via ordered antigenic variation, and disease transmissibility. However, stumpy formation has been lost in most laboratory-adapted trypanosome lines, generating monomorphic parasites that proliferate uncontrolled as slender forms in vitro and in vivo. Nonetheless, these forms are readily amenable to cell culture and high-throughput screening for trypanocidal lead compounds. Here, we have developed and exploited a high-throughput screen for developmental phenotypes using a transgenic monomorphic cell line expressing a reporter under the regulation of gene control signals from the stumpy-specific molecule PAD1. Using a whole-cell fluorescence-based assay to screen over 6,000 small molecules from a kinase-focused compound library, small molecules able to activate stumpy-specific gene expression and proliferation arrest were assayed in a rapid assay format. Independent follow-up validation identified one hit able to induce modest, yet specific, changes in mRNA expression indicative of a partial differentiation to stumpy forms in monomorphs. Further, in pleomorphs this compound induced a stumpy-like phenotype, entailing growth arrest, morphological changes, PAD1 expression, and enhanced differentiation to procyclic forms. This not only provides a potential tool compound for the further understanding of stumpy formation but also demonstrates the use of high-throughput screening in the identification of compounds able to induce specific phenotypes, such as differentiation, in African trypanosomes.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Fenotipo , Proteínas Protozoarias/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Trypanosoma brucei brucei/genética , Proteínas Protozoarias/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/crecimiento & desarrollo , Trypanosoma brucei brucei/patogenicidad , Virulencia/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda