Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(22): 4851-4867.e20, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37848036

RESUMEN

Post-acute sequelae of COVID-19 (PASC, "Long COVID") pose a significant global health challenge. The pathophysiology is unknown, and no effective treatments have been found to date. Several hypotheses have been formulated to explain the etiology of PASC, including viral persistence, chronic inflammation, hypercoagulability, and autonomic dysfunction. Here, we propose a mechanism that links all four hypotheses in a single pathway and provides actionable insights for therapeutic interventions. We find that PASC are associated with serotonin reduction. Viral infection and type I interferon-driven inflammation reduce serotonin through three mechanisms: diminished intestinal absorption of the serotonin precursor tryptophan; platelet hyperactivation and thrombocytopenia, which impacts serotonin storage; and enhanced MAO-mediated serotonin turnover. Peripheral serotonin reduction, in turn, impedes the activity of the vagus nerve and thereby impairs hippocampal responses and memory. These findings provide a possible explanation for neurocognitive symptoms associated with viral persistence in Long COVID, which may extend to other post-viral syndromes.


Asunto(s)
Síndrome Post Agudo de COVID-19 , Serotonina , Humanos , COVID-19/complicaciones , Progresión de la Enfermedad , Inflamación , Síndrome Post Agudo de COVID-19/sangre , Síndrome Post Agudo de COVID-19/patología , Serotonina/sangre , Virosis
2.
Nature ; 630(8015): 174-180, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38811723

RESUMEN

The parasite Cryptosporidium is a leading agent of diarrhoeal disease in young children, and a cause and consequence of chronic malnutrition1,2. There are no vaccines and only limited treatment options3. The parasite infects enterocytes, in which it engages in asexual and sexual replication4, both of which are essential to continued infection and transmission. However, their molecular mechanisms remain largely unclear5. Here we use single-cell RNA sequencing to reveal the gene expression programme of the entire Cryptosporidium parvum life cycle in culture and in infected animals. Diverging from the prevailing model6, we find support for only three intracellular stages: asexual type-I meronts, male gamonts and female gametes. We reveal a highly organized program for the assembly of components at each stage. Dissecting the underlying regulatory network, we identify the transcription factor Myb-M as the earliest determinant of male fate, in an organism that lacks genetic sex determination. Conditional expression of this factor overrides the developmental program and induces widespread maleness, while conditional deletion ablates male development. Both have a profound impact on the infection. A large set of stage-specific genes now provides the opportunity to understand, engineer and disrupt parasite sex and life cycle progression to advance the development of vaccines and treatments.


Asunto(s)
Criptosporidiosis , Cryptosporidium parvum , Regulación de la Expresión Génica , Estadios del Ciclo de Vida , Transcripción Genética , Animales , Femenino , Humanos , Masculino , Ratones , Criptosporidiosis/parasitología , Cryptosporidium parvum/genética , Cryptosporidium parvum/crecimiento & desarrollo , Redes Reguladoras de Genes , Estadios del Ciclo de Vida/genética , Proteínas Proto-Oncogénicas c-myb/genética , Procesos de Determinación del Sexo/genética , Análisis de Expresión Génica de una Sola Célula
3.
Proc Natl Acad Sci U S A ; 121(1): e2313210120, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38147547

RESUMEN

Parasites and their hosts are engaged in reciprocal coevolution that balances competing mechanisms of virulence, resistance, and evasion. This often leads to host specificity, but genomic reassortment between different strains can enable parasites to jump host barriers and conquer new niches. In the apicomplexan parasite Cryptosporidium, genetic exchange has been hypothesized to play a prominent role in adaptation to humans. The sexual lifecycle of the parasite provides a potential mechanism for such exchange; however, the boundaries of Cryptosporidium sex are currently undefined. To explore this experimentally, we established a model for genetic crosses. Drug resistance was engineered using a mutated phenylalanyl tRNA synthetase gene and marking strains with this and the previously used Neo transgene enabled selection of recombinant progeny. This is highly efficient, and genomic recombination is evident and can be continuously monitored in real time by drug resistance, flow cytometry, and PCR mapping. Using this approach, multiple loci can now be modified with ease. We demonstrate that essential genes can be ablated by crossing a Cre recombinase driver strain with floxed strains. We further find that genetic crosses are also feasible between species. Crossing Cryptosporidium parvum, a parasite of cattle and humans, and Cryptosporidium tyzzeri a mouse parasite resulted in progeny with a recombinant genome derived from both species that continues to vigorously replicate sexually. These experiments have important fundamental and translational implications for the evolution of Cryptosporidium and open the door to reverse- and forward-genetic analysis of parasite biology and host specificity.


Asunto(s)
Criptosporidiosis , Cryptosporidium parvum , Cryptosporidium , Cruzamientos Genéticos , Criptosporidiosis/parasitología , Cryptosporidium/genética , Cryptosporidium parvum/genética , Estadios del Ciclo de Vida
4.
EMBO J ; 41(22): e111158, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36245278

RESUMEN

Apicomplexan parasites possess secretory organelles called rhoptries that undergo regulated exocytosis upon contact with the host. This process is essential for the parasitic lifestyle of these pathogens and relies on an exocytic machinery sharing structural features and molecular components with free-living ciliates. However, how the parasites coordinate exocytosis with host interaction is unknown. Here, we performed a Tetrahymena-based transcriptomic screen to uncover novel exocytic factors in Ciliata and conserved in Apicomplexa. We identified membrane-bound proteins, named CRMPs, forming part of a large complex essential for rhoptry secretion and invasion in Toxoplasma. Using cutting-edge imaging tools, including expansion microscopy and cryo-electron tomography, we show that, unlike previously described rhoptry exocytic factors, TgCRMPs are not required for the assembly of the rhoptry secretion machinery and only transiently associate with the exocytic site-prior to the invasion. CRMPs and their partners contain putative host cell-binding domains, and CRMPa shares similarities with GPCR proteins. Collectively our data imply that the CRMP complex acts as a host-molecular sensor to ensure that rhoptry exocytosis occurs when the parasite contacts the host cell.


Asunto(s)
Toxoplasma , Toxoplasma/genética , Toxoplasma/metabolismo , Proteínas Protozoarias/metabolismo , Orgánulos/metabolismo , Exocitosis , Proteínas de la Membrana/metabolismo , Interacciones Huésped-Parásitos
5.
PLoS Pathog ; 20(5): e1011820, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38718306

RESUMEN

The production of IFN-γ is crucial for control of multiple enteric infections, but its impact on intestinal epithelial cells (IEC) is not well understood. Cryptosporidium parasites exclusively infect epithelial cells and the ability of interferons to activate the transcription factor STAT1 in IEC is required for parasite clearance. Here, the use of single cell RNA sequencing to profile IEC during infection revealed an increased proportion of mid-villus enterocytes during infection and induction of IFN-γ-dependent gene signatures that was comparable between uninfected and infected cells. These analyses were complemented by in vivo studies, which demonstrated that IEC expression of the IFN-γ receptor was required for parasite control. Unexpectedly, treatment of Ifng-/- mice with IFN-γ showed the IEC response to this cytokine correlates with a delayed reduction in parasite burden but did not affect parasite development. These data sets provide insight into the impact of IFN-γ on IEC and suggest a model in which IFN-γ signalling to uninfected enterocytes is important for control of Cryptosporidium.


Asunto(s)
Criptosporidiosis , Interferón gamma , Mucosa Intestinal , Ratones Noqueados , Animales , Interferón gamma/metabolismo , Interferón gamma/inmunología , Criptosporidiosis/inmunología , Criptosporidiosis/parasitología , Ratones , Mucosa Intestinal/parasitología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Cryptosporidium , Células Epiteliales/parasitología , Células Epiteliales/metabolismo , Células Epiteliales/inmunología , Enterocitos/parasitología , Enterocitos/metabolismo , Enterocitos/inmunología , Ratones Endogámicos C57BL , Receptor de Interferón gamma , Factor de Transcripción STAT1/metabolismo , Receptores de Interferón/metabolismo , Receptores de Interferón/genética , Transducción de Señal
6.
Genome Res ; 32(1): 203-213, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34764149

RESUMEN

Cryptosporidiosis is a leading cause of waterborne diarrheal disease globally and an important contributor to mortality in infants and the immunosuppressed. Despite its importance, the Cryptosporidium community has only had access to a good, but incomplete, Cryptosporidium parvum IOWA reference genome sequence. Incomplete reference sequences hamper annotation, experimental design, and interpretation. We have generated a new C. parvum IOWA genome assembly supported by Pacific Biosciences (PacBio) and Oxford Nanopore long-read technologies and a new comparative and consistent genome annotation for three closely related species: C. parvum, Cryptosporidium hominis, and Cryptosporidium tyzzeri We made 1926 C. parvum annotation updates based on experimental evidence. They include new transporters, ncRNAs, introns, and altered gene structures. The new assembly and annotation revealed a complete Dnmt2 methylase ortholog. Comparative annotation between C. parvum, C. hominis, and C. tyzzeri revealed that most "missing" orthologs are found, suggesting that the biological differences between the species must result from gene copy number variation, differences in gene regulation, and single-nucleotide variants (SNVs). Using the new assembly and annotation as reference, 190 genes are identified as evolving under positive selection, including many not detected previously. The new C. parvum IOWA reference genome assembly is larger, gap free, and lacks ambiguous bases. This chromosomal assembly recovers all 16 chromosome ends, 13 of which are contiguously assembled. The three remaining chromosome ends are provisionally placed. These ends represent duplication of entire chromosome ends including subtelomeric regions revealing a new level of genome plasticity that will both inform and impact future research.


Asunto(s)
Criptosporidiosis , Cryptosporidium , Criptosporidiosis/genética , Cryptosporidium/genética , Variaciones en el Número de Copia de ADN , Genoma , Humanos , Telómero/genética
7.
PLoS Biol ; 20(4): e3001604, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35436284

RESUMEN

Cryptosporidium is a leading infectious cause of diarrhea around the world associated with waterborne outbreaks, community spread, or zoonotic transmission. The parasite has significant impact on early childhood mortality, and infection is both a consequence and cause of malnutrition and stunting. There is currently no vaccine, and treatment options are very limited. Cryptosporidium is a member of the Apicomplexa, and, as typical for this, protist phylum relies on asexual and sexual reproduction. In contrast to other Apicomplexa, including the malaria parasite Plasmodium, the entire Cryptosporidium life cycle unfolds in a single host in less than 3 days. Here, we establish a model to image life cycle progression in living cells and observe, track, and compare nuclear division of asexual and sexual stage parasites. We establish the length and sequence of the cell cycles of all stages and map the developmental fate of parasites across multiple rounds of invasion and egress. We propose that the parasite executes an intrinsic program of 3 generations of asexual replication, followed by a single generation of sexual stages that is independent of environmental stimuli. We find no evidence for a morphologically distinct intermediate stage (the tetraploid type II meront) but demonstrate direct development of gametes from 8N type I meronts. The progeny of each meront is collectively committed to either asexual or sexual fate, but, importantly, meronts committed to sexual fate give rise to both males and females. We define a Cryptosporidium life cycle matching Tyzzer's original description and inconsistent with the coccidian life cycle now shown in many textbooks.


Asunto(s)
Criptosporidiosis , Cryptosporidium parvum , Cryptosporidium , Animales , Preescolar , Criptosporidiosis/parasitología , Femenino , Células Germinativas , Humanos , Estadios del Ciclo de Vida , Masculino
8.
PLoS Pathog ; 18(5): e1010003, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35584177

RESUMEN

Cryptosporidium is a leading cause of severe diarrhea and diarrheal-related death in children worldwide. As an obligate intracellular parasite, Cryptosporidium relies on intestinal epithelial cells to provide a niche for its growth and survival, but little is known about the contributions that the infected cell makes to this relationship. Here we conducted a genome wide CRISPR/Cas9 knockout screen to discover host genes that influence Cryptosporidium parvum infection and/or host cell survival. Gene enrichment analysis indicated that the host interferon response, glycosaminoglycan (GAG) and glycosylphosphatidylinositol (GPI) anchor biosynthesis are important determinants of susceptibility to C. parvum infection and impact on the viability of host cells in the context of parasite infection. Several of these pathways are linked to parasite attachment and invasion and C-type lectins on the surface of the parasite. Evaluation of transcript and protein induction of innate interferons revealed a pronounced type III interferon response to Cryptosporidium in human cells as well as in mice. Treatment of mice with IFNλ reduced infection burden and protected immunocompromised mice from severe outcomes including death, with effects that required STAT1 signaling in the enterocyte. Initiation of this type III interferon response was dependent on sustained intracellular growth and mediated by the pattern recognition receptor TLR3. We conclude that host cell intrinsic recognition of Cryptosporidium results in IFNλ production critical to early protection against this infection.


Asunto(s)
Criptosporidiosis , Cryptosporidium parvum , Interferones , Receptor Toll-Like 3 , Animales , Criptosporidiosis/genética , Criptosporidiosis/parasitología , Cryptosporidium parvum/genética , Cryptosporidium parvum/inmunología , Diarrea , Interferones/inmunología , Ratones , Receptor Toll-Like 3/inmunología , Interferón lambda
9.
J Immunol ; 209(12): 2261-2268, 2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36469846

RESUMEN

Cryptosporidium is a ubiquitous protozoan parasite that infects gut epithelial cells and causes self-limited diarrhea in immunocompetent individuals. However, in immunocompromised hosts with global defects in T cell function, this infection can result in chronic, life-threatening disease. In addition, there is a subset of individuals with primary immunodeficiencies associated with increased risk for life-threatening cryptosporidiosis. These patients highlight MHC class II expression, CD40-CD40L interactions, NF-κB signaling, and IL-21 as key host factors required for resistance to this enteric pathogen. Understanding which immune deficiencies do (or do not) lead to increased risk for severe Cryptosporidium may reveal mechanisms of parasite restriction and aid in the identification of novel strategies to manage this common pathogen in immunocompetent and deficient hosts.


Asunto(s)
Criptosporidiosis , Cryptosporidium , Síndromes de Inmunodeficiencia , Humanos , Diarrea/complicaciones , Diarrea/parasitología , Huésped Inmunocomprometido
10.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33372132

RESUMEN

The apicomplexan parasite Cryptosporidium infects the intestinal epithelium. While infection is widespread around the world, children in resource-poor settings suffer a disproportionate disease burden. Cryptosporidiosis is a leading cause of diarrheal disease, responsible for mortality and stunted growth in children. CD4 T cells are required to resolve this infection, but powerful innate mechanisms control the parasite prior to the onset of adaptive immunity. Here, we use the natural mouse pathogen Cryptosporidium tyzzeri to demonstrate that the inflammasome plays a critical role in initiating this early response. Mice lacking core inflammasome components, including caspase-1 and apoptosis-associated speck-like protein, show increased parasite burden and caspase 1 deletion solely in enterocytes phenocopies whole-body knockout (KO). This response was fully functional in germfree mice and sufficient to control Cryptosporidium infection. Inflammasome activation leads to the release of IL-18, and mice that lack IL-18 are more susceptible to infection. Treatment of infected caspase 1 KO mice with recombinant IL-18 is remarkably efficient in rescuing parasite control. Notably, NOD-like receptor family pyrin domain containing 6 (NLRP6) was the only NLR required for innate parasite control. Taken together, these data support a model of innate recognition of Cryptosporidium infection through an NLRP6-dependent and enterocyte-intrinsic inflammasome that leads to the release of IL-18 required for parasite control.


Asunto(s)
Criptosporidiosis/inmunología , Enterocitos/metabolismo , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Caspasa 1/metabolismo , Cryptosporidium/fisiología , Enterocitos/inmunología , Interacciones Huésped-Patógeno , Ratones
11.
Nature ; 546(7658): 376-380, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28562588

RESUMEN

Diarrhoeal disease is responsible for 8.6% of global child mortality. Recent epidemiological studies found the protozoan parasite Cryptosporidium to be a leading cause of paediatric diarrhoea, with particularly grave impact on infants and immunocompromised individuals. There is neither a vaccine nor an effective treatment. Here we establish a drug discovery process built on scalable phenotypic assays and mouse models that take advantage of transgenic parasites. Screening a library of compounds with anti-parasitic activity, we identify pyrazolopyridines as inhibitors of Cryptosporidium parvum and Cryptosporidium hominis. Oral treatment with the pyrazolopyridine KDU731 results in a potent reduction in intestinal infection of immunocompromised mice. Treatment also leads to rapid resolution of diarrhoea and dehydration in neonatal calves, a clinical model of cryptosporidiosis that closely resembles human infection. Our results suggest that the Cryptosporidium lipid kinase PI(4)K (phosphatidylinositol-4-OH kinase) is a target for pyrazolopyridines and that KDU731 warrants further preclinical evaluation as a drug candidate for the treatment of cryptosporidiosis.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Criptosporidiosis/tratamiento farmacológico , Criptosporidiosis/parasitología , Cryptosporidium/efectos de los fármacos , Cryptosporidium/enzimología , Pirazoles/farmacología , Piridinas/farmacología , Animales , Animales Recién Nacidos , Bovinos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Huésped Inmunocomprometido , Interferón gamma/deficiencia , Interferón gamma/genética , Masculino , Ratones , Ratones Noqueados , Pirazoles/química , Pirazoles/farmacocinética , Piridinas/química , Piridinas/farmacocinética , Ratas , Ratas Wistar
12.
PLoS Biol ; 17(9): e3000446, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31487278

RESUMEN

Toxoplasma gondii is a remarkably successful protozoan parasite that infects a third of the human population, along with most mammals and birds. However, the sexual portion of the parasite's life cycle is narrowly limited to cats. How parasites distinguish between hosts has long been a mystery. A new study reveals that Toxoplasma identifies cats based on a single fatty acid, linoleic acid. Experimental manipulation of fatty acid metabolism by drug treatment turns a mouse into a cat in the "eye" of the parasite. This new model enables genetic crosses of an important human pathogen without the use of companion animals and opens the door to future discovery.


Asunto(s)
Parásitos , Toxoplasma , Animales , Gatos , Especificidad del Huésped , Humanos , Estadios del Ciclo de Vida , Linoleoil-CoA Desaturasa , Ratones
13.
Proc Natl Acad Sci U S A ; 116(42): 21160-21165, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31570573

RESUMEN

The apicomplexan parasite Cryptosporidium is a leading global cause of severe diarrheal disease and an important contributor to early-childhood mortality. Waterborne outbreaks occur frequently, even in countries with advanced water treatment capabilities, and there is currently no fully effective treatment. Nucleotide pathways are attractive targets for antimicrobial development, and several laboratories are designing inhibitors of these enzymes as potential treatment for Cryptosporidium infections. Here we take advantage of newly available molecular genetics for Cryptosporidium parvum to investigate nucleotide biosynthesis by directed gene ablation. Surprisingly, we found that the parasite tolerates the loss of classical targets including dihydrofolate reductase-thymidylate synthase (DHFR-TS) and inosine monophosphate dehydrogenase (IMPDH). We show that thymidine kinase provides a route to thymidine monophosphate in the absence of DHFR-TS. In contrast, only a single pathway has been identified for C. parvum purine nucleotide salvage. Nonetheless, multiple enzymes in the purine pathway, as well as the adenosine transporter, can be ablated. The resulting mutants are viable under normal conditions but are hypersensitive to inhibition of purine nucleotide synthesis in their host cell. Cryptosporidium might use as-yet undiscovered purine transporters and salvage enzymes; however, genetic and pharmacological experiments led us to conclude that Cryptosporidium imports purine nucleotides from the host cell. The potential for ATP uptake from the host has significant impact on our understanding of parasite energy metabolism given that Cryptosporidium lacks oxidative phosphorylation and glycolytic enzymes are not constitutively expressed throughout the parasite life cycle.


Asunto(s)
Transporte Biológico/fisiología , Criptosporidiosis/metabolismo , Criptosporidiosis/parasitología , Cryptosporidium parvum/genética , Cryptosporidium parvum/metabolismo , Nucleótidos/metabolismo , Purinas/metabolismo , Línea Celular Tumoral , Humanos , IMP Deshidrogenasa/metabolismo , Complejos Multienzimáticos/metabolismo , Tetrahidrofolato Deshidrogenasa/metabolismo , Timidilato Sintasa/metabolismo
14.
Nature ; 523(7561): 477-80, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-26176919

RESUMEN

Recent studies into the global causes of severe diarrhoea in young children have identified the protozoan parasite Cryptosporidium as the second most important diarrhoeal pathogen after rotavirus. Diarrhoeal disease is estimated to be responsible for 10.5% of overall child mortality. Cryptosporidium is also an opportunistic pathogen in the contexts of human immunodeficiency virus (HIV)-caused AIDS and organ transplantation. There is no vaccine and only a single approved drug that provides no benefit for those in gravest danger: malnourished children and immunocompromised patients. Cryptosporidiosis drug and vaccine development is limited by the poor tractability of the parasite, which includes a lack of systems for continuous culture, facile animal models, and molecular genetic tools. Here we describe an experimental framework to genetically modify this important human pathogen. We established and optimized transfection of C. parvum sporozoites in tissue culture. To isolate stable transgenics we developed a mouse model that delivers sporozoites directly into the intestine, a Cryptosporidium clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 system, and in vivo selection for aminoglycoside resistance. We derived reporter parasites suitable for in vitro and in vivo drug screening, and we evaluated the basis of drug susceptibility by gene knockout. We anticipate that the ability to genetically engineer this parasite will be transformative for Cryptosporidium research. Genetic reporters will provide quantitative correlates for disease, cure and protection, and the role of parasite genes in these processes is now open to rigorous investigation.


Asunto(s)
Criptosporidiosis/parasitología , Cryptosporidium parvum/genética , Diarrea/parasitología , Ingeniería Genética/métodos , Aminoglicósidos/farmacología , Animales , Antimaláricos/farmacología , Sistemas CRISPR-Cas , Línea Celular , Criptosporidiosis/complicaciones , Cryptosporidium parvum/enzimología , Cryptosporidium parvum/crecimiento & desarrollo , Diarrea/complicaciones , Evaluación Preclínica de Medicamentos , Resistencia a Medicamentos , Femenino , Eliminación de Gen , Técnicas de Inactivación de Genes , Genes Reporteros , Humanos , Intestinos/parasitología , Ratones , Modelos Animales , Esporozoítos , Timidina Quinasa/deficiencia , Timidina Quinasa/genética , Transfección/métodos , Trimetoprim/farmacología
15.
Int J Mol Sci ; 22(12)2021 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-34204357

RESUMEN

Heme biosynthesis is essential for almost all living organisms. Despite its conserved function, the pathway's enzymes can be located in a remarkable diversity of cellular compartments in different organisms. This location does not always reflect their evolutionary origins, as might be expected from the history of their acquisition through endosymbiosis. Instead, the final subcellular localization of the enzyme reflects multiple factors, including evolutionary origin, demand for the product, availability of the substrate, and mechanism of pathway regulation. The biosynthesis of heme in the apicomonad Chromera velia follows a chimeric pathway combining heme elements from the ancient algal symbiont and the host. Computational analyses using different algorithms predict complex targeting patterns, placing enzymes in the mitochondrion, plastid, endoplasmic reticulum, or the cytoplasm. We employed heterologous reporter gene expression in the apicomplexan parasite Toxoplasma gondii and the diatom Phaeodactylum tricornutum to experimentally test these predictions. 5-aminolevulinate synthase was located in the mitochondria in both transfection systems. In T. gondii, the two 5-aminolevulinate dehydratases were located in the cytosol, uroporphyrinogen synthase in the mitochondrion, and the two ferrochelatases in the plastid. In P. tricornutum, all remaining enzymes, from ALA-dehydratase to ferrochelatase, were placed either in the endoplasmic reticulum or in the periplastidial space.


Asunto(s)
Alveolados/fisiología , Apicomplexa/metabolismo , Diatomeas/metabolismo , Hemo/metabolismo , Redes y Vías Metabólicas , Secuencia de Aminoácidos , Transporte Biológico , Evolución Molecular , Regulación Enzimológica de la Expresión Génica , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
16.
PLoS Pathog ; 14(2): e1006836, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29470517

RESUMEN

Apicomplexan parasites are global killers, being the causative agents of diseases like toxoplasmosis and malaria. These parasites are known to be hypersensitive to redox imbalance, yet little is understood about the cellular roles of their various redox regulators. The apicoplast, an essential plastid organelle, is a verified apicomplexan drug target. Nuclear-encoded apicoplast proteins traffic through the ER and multiple apicoplast sub-compartments to their place of function. We propose that thioredoxins contribute to the control of protein trafficking and of protein function within these apicoplast compartments. We studied the role of two Toxoplasma gondii apicoplast thioredoxins (TgATrx), both essential for parasite survival. By describing the cellular phenotypes of the conditional depletion of either of these redox regulated enzymes we show that each of them contributes to a different apicoplast biogenesis pathway. We provide evidence for TgATrx1's involvement in ER to apicoplast trafficking and TgATrx2 in the control of apicoplast gene expression components. Substrate pull-down further recognizes gene expression factors that interact with TgATrx2. We use genetic complementation to demonstrate that the function of both TgATrxs is dependent on their disulphide exchange activity. Finally, TgATrx2 is divergent from human thioredoxins. We demonstrate its activity in vitro thus providing scope for drug screening. Our study represents the first functional characterization of thioredoxins in Toxoplasma, highlights the importance of redox regulation of apicoplast functions and provides new tools to study redox biology in these parasites.


Asunto(s)
Apicoplastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Biogénesis de Organelos , Tiorredoxinas/metabolismo , Toxoplasma/fisiología , Secuencia de Aminoácidos , Biomarcadores/metabolismo , Secuencia Conservada , Evolución Molecular , Técnicas de Silenciamiento del Gen , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Mutación , Filogenia , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Multimerización de Proteína , Transporte de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Tiorredoxinas/química , Tiorredoxinas/genética , Toxoplasma/citología , Toxoplasma/crecimiento & desarrollo
17.
J Infect Dis ; 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38813921
18.
Annu Rev Microbiol ; 67: 271-89, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23808340

RESUMEN

Plasmodium and Toxoplasma are genera of apicomplexan parasites that infect millions of people each year. The former causes malaria, and the latter causes neurotropic infections associated with a weakened or developing immune system. These parasites harbor a peculiar organelle, the apicoplast. The apicoplast is the product of an ancient endosymbiosis between a heterotrophic and a photosynthetic protist. We explore the cellular and molecular mechanisms that enabled a stable union of two previously independent organisms. These include the exchange of metabolites, transfer of genes, transport of proteins, and overall coordination of biogenesis and proliferation. These mechanisms are still active today and can be exploited to treat parasite infection. They were shaped by the dramatic changes that occurred in the evolution of the phylum Apicomplexa--including the gain and loss of photosynthesis, adaptation to symbiosis and parasitism, and the explosion of animal diversity-that ultimately provided an aquatic alga access to every biotope on this planet.


Asunto(s)
Apicomplexa/metabolismo , Apicoplastos/metabolismo , Parásitos/metabolismo , Rhodophyta/metabolismo , Animales , Apicomplexa/genética , Apicoplastos/genética , Evolución Biológica , Humanos , Parásitos/genética , Infecciones por Protozoos/parasitología , Rhodophyta/genética
19.
Immunity ; 31(2): 342-55, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19699173

RESUMEN

Memory T cells circulate through lymph nodes where they are poised to respond rapidly upon re-exposure to a pathogen; however, the dynamics of memory T cell, antigen-presenting cell, and pathogen interactions during recall responses are largely unknown. We used a mouse model of infection with the intracellular protozoan parasite, Toxoplasma gondii, in conjunction with two-photon microscopy, to address this question. After challenge, memory T cells migrated more rapidly than naive T cells, relocalized toward the subcapsular sinus (SCS) near invaded macrophages, and engaged in prolonged interactions with infected cells. Parasite invasion of T cells occurred by direct transfer of the parasite from the target cell into the T cell and corresponded to an antigen-specific increase in the rate of T cell invasion. Our results provide insight into cellular interactions during recall responses and suggest a mechanism of pathogen subversion of the immune response.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Interacciones Huésped-Parásitos/inmunología , Memoria Inmunológica , Ganglios Linfáticos/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/parasitología , Antígeno CD11c/inmunología , Movimiento Celular/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/parasitología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/parasitología , Toxoplasma/inmunología , Toxoplasmosis/inmunología
20.
PLoS Biol ; 13(3): e1002093, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25734885

RESUMEN

Apicomplexan parasites can change fundamental features of cell division during their life cycles, suspending cytokinesis when needed and changing proliferative scale in different hosts and tissues. The structural and molecular basis for this remarkable cell cycle flexibility is not fully understood, although the centrosome serves a key role in determining when and how much replication will occur. Here we describe the discovery of multiple replicating core complexes with distinct protein composition and function in the centrosome of Toxoplasma gondii. An outer core complex distal from the nucleus contains the TgCentrin1/TgSfi1 protein pair, along with the cartwheel protein TgSas-6 and a novel Aurora-related kinase, while an inner core closely aligned with the unique spindle pole (centrocone) holds distant orthologs of the CEP250/C-Nap protein family. This outer/inner spatial relationship of centrosome cores is maintained throughout the cell cycle. When in metaphase, the duplicated cores align to opposite sides of the kinetochores in a linear array. As parasites transition into S phase, the cores sequentially duplicate, outer core first and inner core second, ensuring that each daughter parasite inherits one copy of each type of centrosome core. A key serine/threonine kinase distantly related to the MAPK family is localized to the centrosome, where it restricts core duplication to once per cycle and ensures the proper formation of new daughter parasites. Genetic analysis of the outer core in a temperature-sensitive mutant demonstrated this core functions primarily in cytokinesis. An inhibition of ts-TgSfi1 function at high temperature caused the loss of outer cores and a severe block to budding, while at the same time the inner core amplified along with the unique spindle pole, indicating the inner core and spindle pole are independent and co-regulated. The discovery of a novel bipartite organization in the parasite centrosome that segregates the functions of karyokinesis and cytokinesis provides an explanation for how cell cycle flexibility is achieved in apicomplexan life cycles.


Asunto(s)
Proteínas de Ciclo Celular/genética , División del Núcleo Celular , Centrosoma/metabolismo , Citocinesis , Proteínas Protozoarias/genética , Toxoplasma/genética , Aurora Quinasas/genética , Aurora Quinasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Centrosoma/ultraestructura , Medios de Cultivo , Fibroblastos/parasitología , Fibroblastos/patología , Regulación de la Expresión Génica , Humanos , Cinetocoros/metabolismo , Cinetocoros/ultraestructura , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Cultivo Primario de Células , Proteínas Protozoarias/metabolismo , Transducción de Señal , Temperatura , Toxoplasma/metabolismo , Toxoplasma/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA