Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 595(7865): 96-100, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34040257

RESUMEN

Trypanosomes are protozoan parasites that cause infectious diseases, including African trypanosomiasis (sleeping sickness) in humans and nagana in economically important livestock1,2. An effective vaccine against trypanosomes would be an important control tool, but the parasite has evolved sophisticated immunoprotective mechanisms-including antigenic variation3-that present an apparently insurmountable barrier to vaccination. Here we show, using a systematic genome-led vaccinology approach and a mouse model of Trypanosoma vivax infection4, that protective invariant subunit vaccine antigens can be identified. Vaccination with a single recombinant protein comprising the extracellular region of a conserved cell-surface protein that is localized to the flagellum membrane (which we term 'invariant flagellum antigen from T. vivax') induced long-lasting protection. Immunity was passively transferred with immune serum, and recombinant monoclonal antibodies to this protein could induce sterile protection and revealed several mechanisms of antibody-mediated immunity, including a major role for complement. Our discovery identifies a vaccine candidate for an important parasitic disease that has constrained socioeconomic development in countries in sub-Saharan Africa5, and provides evidence that highly protective vaccines against trypanosome infections can be achieved.


Asunto(s)
Antígenos de Protozoos/inmunología , Vacunas Antiprotozoos/inmunología , Trypanosoma vivax/inmunología , Tripanosomiasis Africana/inmunología , Tripanosomiasis Africana/prevención & control , Animales , Antígenos de Protozoos/química , Proteínas del Sistema Complemento/inmunología , Secuencia Conservada/inmunología , Modelos Animales de Enfermedad , Femenino , Flagelos/química , Flagelos/inmunología , Ratones , Ratones Endogámicos BALB C , Vacunas Antiprotozoos/química , Factores de Tiempo , Trypanosoma vivax/química , Trypanosoma vivax/citología , Tripanosomiasis Africana/parasitología , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología
2.
Circ Res ; 134(10): 1306-1326, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38533639

RESUMEN

BACKGROUND: Ventricular arrhythmias (VAs) demonstrate a prominent day-night rhythm, commonly presenting in the morning. Transcriptional rhythms in cardiac ion channels accompany this phenomenon, but their role in the morning vulnerability to VAs and the underlying mechanisms are not understood. We investigated the recruitment of transcription factors that underpins transcriptional rhythms in ion channels and assessed whether this mechanism was pertinent to the heart's intrinsic diurnal susceptibility to VA. METHODS AND RESULTS: Assay for transposase-accessible chromatin with sequencing performed in mouse ventricular myocyte nuclei at the beginning of the animals' inactive (ZT0) and active (ZT12) periods revealed differentially accessible chromatin sites annotating to rhythmically transcribed ion channels and distinct transcription factor binding motifs in these regions. Notably, motif enrichment for the glucocorticoid receptor (GR; transcriptional effector of corticosteroid signaling) in open chromatin profiles at ZT12 was observed, in line with the well-recognized ZT12 peak in circulating corticosteroids. Molecular, electrophysiological, and in silico biophysically-detailed modeling approaches demonstrated GR-mediated transcriptional control of ion channels (including Scn5a underlying the cardiac Na+ current, Kcnh2 underlying the rapid delayed rectifier K+ current, and Gja1 responsible for electrical coupling) and their contribution to the day-night rhythm in the vulnerability to VA. Strikingly, both pharmacological block of GR and cardiomyocyte-specific genetic knockout of GR blunted or abolished ion channel expression rhythms and abolished the ZT12 susceptibility to pacing-induced VA in isolated hearts. CONCLUSIONS: Our study registers a day-night rhythm in chromatin accessibility that accompanies diurnal cycles in ventricular myocytes. Our approaches directly implicate the cardiac GR in the myocyte excitability rhythm and mechanistically link the ZT12 surge in glucocorticoids to intrinsic VA propensity at this time.


Asunto(s)
Ritmo Circadiano , Miocitos Cardíacos , Receptores de Glucocorticoides , Animales , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/genética , Ratones , Miocitos Cardíacos/metabolismo , Masculino , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/genética , Ratones Endogámicos C57BL , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Conexina 43/metabolismo , Conexina 43/genética , Ratones Noqueados , Potenciales de Acción
3.
Compr Rev Food Sci Food Saf ; 23(2): e13289, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38343297

RESUMEN

Whey protein denaturation and aggregation have long been areas of research interest to the dairy industry, having significant implications for process performance and final product functionality and quality. As such, a significant number of analytical techniques have been developed or adapted to assess and characterize levels of whey protein denaturation and aggregation, to either maximize processing efficiency or create products with enhanced functionality (both technological and biological). This review aims to collate and critique these approaches based on their analytical principles and outline their application for the assessment of denaturation and aggregation. This review also provides insights into recent developments in process analytical technologies relating to whey protein denaturation and aggregation, whereby some of the analytical methods have been adapted to enable measurements in-line. Developments in this area will enable more live, in-process data to be generated, which will subsequently allow more adaptive processing, enabling improved product quality and processing efficiency. Along with the applicability of these techniques for the assessment of whey protein denaturation and aggregation, limitations are also presented to help assess the suitability of each analytical technique for specific areas of interest.


Asunto(s)
Suero Lácteo , Proteína de Suero de Leche , Desnaturalización Proteica , Concentración de Iones de Hidrógeno
4.
Nature ; 533(7604): 543-546, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27144353

RESUMEN

Our intestinal microbiota harbours a diverse bacterial community required for our health, sustenance and wellbeing. Intestinal colonization begins at birth and climaxes with the acquisition of two dominant groups of strict anaerobic bacteria belonging to the Firmicutes and Bacteroidetes phyla. Culture-independent, genomic approaches have transformed our understanding of the role of the human microbiome in health and many diseases. However, owing to the prevailing perception that our indigenous bacteria are largely recalcitrant to culture, many of their functions and phenotypes remain unknown. Here we describe a novel workflow based on targeted phenotypic culturing linked to large-scale whole-genome sequencing, phylogenetic analysis and computational modelling that demonstrates that a substantial proportion of the intestinal bacteria are culturable. Applying this approach to healthy individuals, we isolated 137 bacterial species from characterized and candidate novel families, genera and species that were archived as pure cultures. Whole-genome and metagenomic sequencing, combined with computational and phenotypic analysis, suggests that at least 50-60% of the bacterial genera from the intestinal microbiota of a healthy individual produce resilient spores, specialized for host-to-host transmission. Our approach unlocks the human intestinal microbiota for phenotypic analysis and reveals how a marked proportion of oxygen-sensitive intestinal bacteria can be transmitted between individuals, affecting microbiota heritability.


Asunto(s)
Bacterias/clasificación , Bacterias/crecimiento & desarrollo , Técnicas de Tipificación Bacteriana , Microbioma Gastrointestinal/fisiología , Anaerobiosis , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/aislamiento & purificación , Técnicas de Cultivo de Célula , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Genoma Bacteriano/genética , Salud , Humanos , Metagenoma/genética , Metagenómica , Oxígeno/metabolismo , Oxígeno/farmacología , Fenotipo , Filogenia , Análisis de Secuencia de ADN , Especificidad de la Especie , Esporas Bacterianas/clasificación , Esporas Bacterianas/efectos de los fármacos , Esporas Bacterianas/crecimiento & desarrollo
5.
PLoS Pathog ; 15(1): e1007265, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30640950

RESUMEN

The whipworm Trichuris trichiura is a soil-transmitted helminth that dwells in the epithelium of the caecum and proximal colon of their hosts causing the human disease, trichuriasis. Trichuriasis is characterized by colitis attributed to the inflammatory response elicited by the parasite while tunnelling through intestinal epithelial cells (IECs). The IL-10 family of receptors, comprising combinations of subunits IL-10Rα, IL-10Rß, IL-22Rα and IL-28Rα, modulates intestinal inflammatory responses. Here we carefully dissected the role of these subunits in the resistance of mice to infection with T. muris, a mouse model of the human whipworm T. trichiura. Our findings demonstrate that whilst IL-22Rα and IL-28Rα are dispensable in the host response to whipworms, IL-10 signalling through IL-10Rα and IL-10Rß is essential to control caecal pathology, worm expulsion and survival during T. muris infections. We show that deficiency of IL-10, IL-10Rα and IL-10Rß results in dysbiosis of the caecal microbiota characterised by expanded populations of opportunistic bacteria of the families Enterococcaceae and Enterobacteriaceae. Moreover, breakdown of the epithelial barrier after whipworm infection in IL-10, IL-10Rα and IL-10Rß-deficient mice, allows the translocation of these opportunistic pathogens or their excretory products to the liver causing organ failure and lethal disease. Importantly, bone marrow chimera experiments indicate that signalling through IL-10Rα and IL-10Rß in haematopoietic cells, but not IECs, is crucial to control worm expulsion and immunopathology. These findings are supported by worm expulsion upon infection of conditional mutant mice for the IL-10Rα on IECs. Our findings emphasize the pivotal and complex role of systemic IL-10Rα signalling on immune cells in promoting microbiota homeostasis and maintaining the intestinal epithelial barrier, thus preventing immunopathology during whipworm infections.


Asunto(s)
Interleucina-10/metabolismo , Receptores de Interleucina-10/metabolismo , Trichuris/inmunología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/inmunología , Homeostasis , Interleucinas/metabolismo , Intestinos/microbiología , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Tricuriasis/inmunología , Trichuris/parasitología , Interleucina-22
6.
Proc Natl Acad Sci U S A ; 115(40): 10118-10123, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30217896

RESUMEN

Intestinal epithelial cells (IECs) play a key role in regulating immune responses and controlling infection. However, the direct role of IECs in restricting pathogens remains incompletely understood. Here, we provide evidence that IL-22 primed intestinal organoids derived from healthy human induced pluripotent stem cells (hIPSCs) to restrict Salmonella enterica serovar Typhimurium SL1344 infection. A combination of transcriptomics, bacterial invasion assays, and imaging suggests that IL-22-induced antimicrobial activity is driven by increased phagolysosomal fusion in IL-22-pretreated cells. The antimicrobial phenotype was absent in hIPSCs derived from a patient harboring a homozygous mutation in the IL10RB gene that inactivates the IL-22 receptor but was restored by genetically complementing the IL10RB deficiency. This study highlights a mechanism through which the IL-22 pathway facilitates the human intestinal epithelium to control microbial infection.


Asunto(s)
Células Epiteliales/inmunología , Células Madre Pluripotentes Inducidas/inmunología , Interleucinas/inmunología , Mucosa Intestinal/inmunología , Fagosomas/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Células Epiteliales/microbiología , Células Epiteliales/patología , Humanos , Células Madre Pluripotentes Inducidas/microbiología , Células Madre Pluripotentes Inducidas/patología , Subunidad beta del Receptor de Interleucina-10/genética , Subunidad beta del Receptor de Interleucina-10/inmunología , Subunidad alfa del Receptor de Interleucina-21/genética , Subunidad alfa del Receptor de Interleucina-21/inmunología , Interleucinas/genética , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Fagosomas/genética , Fagosomas/microbiología , Fagosomas/patología , Infecciones por Salmonella/genética , Infecciones por Salmonella/patología , Salmonella typhimurium/genética , Interleucina-22
7.
Genes Dev ; 27(10): 1198-215, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23699412

RESUMEN

Fertilization is a crucial yet poorly characterized event in eukaryotes. Our previous discovery that the broadly conserved protein HAP2 (GCS1) functioned in gamete membrane fusion in the unicellular green alga Chlamydomonas and the malaria pathogen Plasmodium led us to exploit the rare biological phenomenon of isogamy in Chlamydomonas in a comparative transcriptomics strategy to uncover additional conserved sexual reproduction genes. All previously identified Chlamydomonas fertilization-essential genes fell into related clusters based on their expression patterns. Out of several conserved genes in a minus gamete cluster, we focused on Cre06.g280600, an ortholog of the fertilization-related Arabidopsis GEX1. Gene disruption, cell biological, and immunolocalization studies show that CrGEX1 functions in nuclear fusion in Chlamydomonas. Moreover, CrGEX1 and its Plasmodium ortholog, PBANKA_113980, are essential for production of viable meiotic progeny in both organisms and thus for mosquito transmission of malaria. Remarkably, we discovered that the genes are members of a large, previously unrecognized family whose first-characterized member, KAR5, is essential for nuclear fusion during yeast sexual reproduction. Our comparative transcriptomics approach provides a new resource for studying sexual development and demonstrates that exploiting the data can lead to the discovery of novel biology that is conserved across distant taxa.


Asunto(s)
Chlamydomonas/genética , Hongos/genética , Genes Esenciales , Membrana Nuclear/metabolismo , Proteínas Nucleares/clasificación , Plasmodium/genética , Vertebrados/genética , Animales , Proteínas de Arabidopsis/clasificación , Proteínas de Arabidopsis/metabolismo , Fertilización/genética , Hongos/crecimiento & desarrollo , Perfilación de la Expresión Génica , Meiosis , Proteínas de la Membrana/clasificación , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Plantas/genética , Reproducción/genética , Proteínas de Saccharomyces cerevisiae/clasificación , Proteínas de Saccharomyces cerevisiae/metabolismo , Transcriptoma/genética
8.
Mol Microbiol ; 112(6): 1831-1846, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31556164

RESUMEN

The discovery of a Salmonella-targeting phage from the waterways of the United Kingdom provided an opportunity to address the mechanism by which Chi-like bacteriophage (phage) engages with bacterial flagellae. The long tail fibre seen on Chi-like phages has been proposed to assist the phage particle in docking to a host cell flagellum, but the identity of the protein that generates this fibre was unknown. We present the results from genome sequencing of this phage, YSD1, confirming its close relationship to the original Chi phage and suggesting candidate proteins to form the tail structure. Immunogold labelling in electron micrographs revealed that YSD1_22 forms the main shaft of the tail tube, while YSD1_25 forms the distal part contributing to the tail spike complex. The long curling tail fibre is formed by the protein YSD1_29, and treatment of phage with the antibodies that bind YSD1_29 inhibits phage infection of Salmonella. The host range for YSD1 across Salmonella serovars is broad, but not comprehensive, being limited by antigenic features of the flagellin subunits that make up the Salmonella flagellum, with which YSD1_29 engages to initiate infection.


Asunto(s)
Flagelos/genética , Fagos de Salmonella/genética , Fagos de Salmonella/aislamiento & purificación , Bacteriófagos/genética , ADN Viral/genética , Flagelos/metabolismo , Flagelos/fisiología , Genoma Viral/genética , Especificidad del Huésped , Filogenia , Fagos de Salmonella/metabolismo , Salmonella typhi/genética , Salmonella typhi/metabolismo , Análisis de Secuencia de ADN/métodos , Reino Unido
9.
Nature ; 508(7497): 483-7, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24739963

RESUMEN

Fertilization occurs when sperm and egg recognize each other and fuse to form a new, genetically distinct organism. The molecular basis of sperm-egg recognition is unknown, but is likely to require interactions between receptor proteins displayed on their surface. Izumo1 is an essential sperm cell-surface protein, but its receptor on the egg has not been described. Here we identify folate receptor 4 (Folr4) as the receptor for Izumo1 on the mouse egg, and propose to rename it Juno. We show that the Izumo1-Juno interaction is conserved within several mammalian species, including humans. Female mice lacking Juno are infertile and Juno-deficient eggs do not fuse with normal sperm. Rapid shedding of Juno from the oolemma after fertilization suggests a mechanism for the membrane block to polyspermy, ensuring eggs normally fuse with just a single sperm. Our discovery of an essential receptor pair at the nexus of conception provides opportunities for the rational development of new fertility treatments and contraceptives.


Asunto(s)
Fertilización/fisiología , Inmunoglobulinas/metabolismo , Proteínas de la Membrana/metabolismo , Óvulo/metabolismo , Receptores de Superficie Celular/metabolismo , Espermatozoides/metabolismo , Animales , Secuencia Conservada , Evolución Molecular , Femenino , Fertilidad/genética , Fertilización/genética , Genes Esenciales , Glicosilfosfatidilinositoles/metabolismo , Humanos , Infertilidad Femenina/genética , Masculino , Mamíferos , Ratones , Oocitos/citología , Oocitos/metabolismo , Óvulo/citología , Partenogénesis , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Inyecciones de Esperma Intracitoplasmáticas , Factores de Tiempo
10.
Proc Biol Sci ; 286(1900): 20182025, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30966987

RESUMEN

The sixth global cholera pandemic lasted from 1899 to 1923. However, despite widespread fear of the disease and of its negative effects on troop morale, very few soldiers in the British Expeditionary Forces contracted cholera between 1914 and 1918. Here, we have revived and sequenced the genome of NCTC 30, a 102-year-old Vibrio cholerae isolate, which we believe is the oldest publicly available live V. cholerae strain in existence. NCTC 30 was isolated in 1916 from a British soldier convalescent in Egypt. We found that this strain does not encode cholera toxin, thought to be necessary to cause cholera, and is not part of V. cholerae lineages responsible for the pandemic disease. We also show that NCTC 30, which predates the introduction of penicillin-based antibiotics, harbours a functional ß-lactamase antibiotic resistance gene. Our data corroborate and provide molecular explanations for previous phenotypic studies of NCTC 30 and provide a new high-quality genome sequence for historical, non-pandemic V. cholerae.


Asunto(s)
Cólera/historia , Genoma Bacteriano , Vibrio cholerae/genética , Cólera/microbiología , Historia del Siglo XX , Análisis de Secuencia de ADN , Primera Guerra Mundial
11.
Int J Med Microbiol ; 308(7): 890-898, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29937391

RESUMEN

Bacteriophages play an important role in the evolution of bacterial pathogens. A phage-mediated transfer of stx-genes to atypical enteropathogenic E. coli (aEPEC) which are prevalent in different hosts, would convert them to enterohemorrhagic E. coli (EHEC). We decided to confirm this hypothesis experimentally to provide conclusive evidence that aEPEC isolated from different mammalian hosts are indeed progenitors of typical EHEC which gain the ability to produce Shiga-Toxin by lysogeny with stx-converting bacteriophages, utilizing the model phage Φ3538 Δstx2::cat. We applied a modified in vitro plaque-assay, using a high titer of a bacteriophage carrying a deletion in the stx2 gene (Φ3538 Δstx2::cat) to increase the detection of lysogenic conversion events. Three wild-type aEPEC strains were chosen as acceptor strains: the murine aEPEC-strain IMT14505 (sequence type (ST)28, serotype Ont:H6), isolated from a striped field mouse (Apodemus agrarius) in the surrounding of a cattle shed, and the human aEPEC-strain 910#00 (ST28, Ont:H6). The close genomic relationship of both strains implies a high zoonotic potential. A third strain, the bovine aEPEC IMT19981, was of serotype O26:H11 and ST21 (STC29). All three aEPEC were successfully lysogenized with phage Φ3538 Δstx2::cat. Integration of the bacteriophage DNA into the aEPEC host genomes was confirmed by amplification of chloramphenicol transferase (cat) marker gene and by Southern-Blot hybridization. Analysis of the whole genome sequence of each of the three lysogens showed that the bacteriophage was integrated into the known tRNA integration site argW, which is highly variable among E. coli. In conclusion, the successful lysogenic conversion of aEPEC with a stx-phage in vitro underlines the important role of aEPEC as progenitors of EHEC. Given the high prevalence and the wide host range of aEPEC acceptors, their high risk of zoonotic transmission should be recognized in infection control measures.


Asunto(s)
Bacteriófagos/genética , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/patogenicidad , Lisogenia/genética , Toxina Shiga/genética , Animales , Bacteriófagos/crecimiento & desarrollo , Bovinos , Escherichia coli Enteropatógena/aislamiento & purificación , Infecciones por Escherichia coli/microbiología , Heces/microbiología , Genoma Viral/genética , Humanos , Ratones
12.
Nature ; 484(7395): 519-23, 2012 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-22446628

RESUMEN

The 2009 H1N1 influenza pandemic showed the speed with which a novel respiratory virus can spread and the ability of a generally mild infection to induce severe morbidity and mortality in a subset of the population. Recent in vitro studies show that the interferon-inducible transmembrane (IFITM) protein family members potently restrict the replication of multiple pathogenic viruses. Both the magnitude and breadth of the IFITM proteins' in vitro effects suggest that they are critical for intrinsic resistance to such viruses, including influenza viruses. Using a knockout mouse model, we now test this hypothesis directly and find that IFITM3 is essential for defending the host against influenza A virus in vivo. Mice lacking Ifitm3 display fulminant viral pneumonia when challenged with a normally low-pathogenicity influenza virus, mirroring the destruction inflicted by the highly pathogenic 1918 'Spanish' influenza. Similar increased viral replication is seen in vitro, with protection rescued by the re-introduction of Ifitm3. To test the role of IFITM3 in human influenza virus infection, we assessed the IFITM3 alleles of individuals hospitalized with seasonal or pandemic influenza H1N1/09 viruses. We find that a statistically significant number of hospitalized subjects show enrichment for a minor IFITM3 allele (SNP rs12252-C) that alters a splice acceptor site, and functional assays show the minor CC genotype IFITM3 has reduced influenza virus restriction in vitro. Together these data reveal that the action of a single intrinsic immune effector, IFITM3, profoundly alters the course of influenza virus infection in mouse and humans.


Asunto(s)
Virus de la Influenza A/patogenicidad , Proteínas de la Membrana/metabolismo , Infecciones por Orthomyxoviridae/mortalidad , Proteínas de Unión al ARN/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Citocinas/inmunología , Inglaterra/epidemiología , Eliminación de Gen , Humanos , Subtipo H1N1 del Virus de la Influenza A/clasificación , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H3N2 del Virus de la Influenza A/clasificación , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Virus de la Influenza A/clasificación , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza B/clasificación , Virus de la Influenza B/crecimiento & desarrollo , Virus de la Influenza B/patogenicidad , Gripe Humana/complicaciones , Gripe Humana/epidemiología , Gripe Humana/mortalidad , Gripe Humana/virología , Leucocitos/inmunología , Pulmón/patología , Pulmón/virología , Proteínas de la Membrana/química , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/patología , Neumonía Viral/etiología , Neumonía Viral/patología , Neumonía Viral/prevención & control , Polimorfismo de Nucleótido Simple/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Escocia/epidemiología , Replicación Viral
13.
J Biol Chem ; 291(27): 14285-14299, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27226583

RESUMEN

Diversity at pathogen genetic loci can be driven by host adaptive immune selection pressure and may reveal proteins important for parasite biology. Population-based genome sequencing of Plasmodium falciparum, the parasite responsible for the most severe form of malaria, has highlighted two related polymorphic genes called dblmsp and dblmsp2, which encode Duffy binding-like (DBL) domain-containing proteins located on the merozoite surface but whose function remains unknown. Using recombinant proteins and transgenic parasites, we show that DBLMSP and DBLMSP2 directly and avidly bind human IgM via their DBL domains. We used whole genome sequence data from over 400 African and Asian P. falciparum isolates to show that dblmsp and dblmsp2 exhibit extreme protein polymorphism in their DBL domain, with multiple variants of two major allelic classes present in every population tested. Despite this variability, the IgM binding function was retained across diverse sequence representatives. Although this interaction did not seem to have an effect on the ability of the parasite to invade red blood cells, binding of DBLMSP and DBLMSP2 to IgM inhibited the overall immunoreactivity of these proteins to IgG from patients who had been exposed to the parasite. This suggests that IgM binding might mask these proteins from the host humoral immune system.


Asunto(s)
Antígenos de Protozoos/metabolismo , Inmunoglobulina M/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Humanos , Unión Proteica
14.
Infect Immun ; 85(9)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28674031

RESUMEN

The ST313 pathovar of Salmonella enterica serovar Typhimurium contributes to a high burden of invasive disease among African infants and HIV-infected adults. It is characterized by genome degradation (loss of coding capacity) and has increased resistance to antibody-dependent complement-mediated killing compared with enterocolitis-causing strains of S Typhimurium. Vaccination is an attractive disease-prevention strategy, and leading candidates focus on the induction of bactericidal antibodies. Antibody-resistant strains arising through further gene deletion could compromise such a strategy. Exposing a saturating transposon insertion mutant library of S Typhimurium to immune serum identified a repertoire of S Typhimurium genes that, when interrupted, result in increased resistance to serum killing. These genes included several involved in bacterial envelope biogenesis, protein translocation, and metabolism. We generated defined mutant derivatives using S Typhimurium SL1344 as the host. Based on their initial levels of enhanced resistance to killing, yfgA and sapA mutants were selected for further characterization. The S Typhimurium yfgA mutant lost the characteristic Salmonella rod-shaped appearance, exhibited increased sensitivity to osmotic and detergent stress, lacked very long lipopolysaccharide, was unable to invade enterocytes, and demonstrated decreased ability to infect mice. In contrast, the S Typhimurium sapA mutants had similar sensitivity to osmotic and detergent stress and lipopolysaccharide profile and an increased ability to infect enterocytes compared with the wild type, but it had no increased ability to cause in vivo infection. These findings indicate that increased resistance to antibody-dependent complement-mediated killing secondary to genetic deletion is not necessarily accompanied by increased virulence and suggest the presence of different mechanisms of antibody resistance.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Anticuerpos Antibacterianos/inmunología , Proteínas Bacterianas/metabolismo , Actividad Bactericida de la Sangre , Proteínas del Sistema Complemento/inmunología , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Transportadoras de Casetes de Unión a ATP/genética , Animales , Proteínas Bacterianas/genética , Elementos Transponibles de ADN , Femenino , Técnicas de Inactivación de Genes , Ratones Endogámicos C57BL , Mutagénesis Insercional , Salmonella typhimurium/fisiología , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
15.
PLoS Pathog ; 11(3): e1004749, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25794007

RESUMEN

Shigella is the leading cause for dysentery worldwide. Together with several virulence factors employed for invasion, the presence and length of the O antigen (OAg) of the lipopolysaccharide (LPS) plays a key role in pathogenesis. S. flexneri 2a has a bimodal OAg chain length distribution regulated in a growth-dependent manner, whereas S. sonnei LPS comprises a monomodal OAg. Here we reveal that S. sonnei, but not S. flexneri 2a, possesses a high molecular weight, immunogenic group 4 capsule, characterized by structural similarity to LPS OAg. We found that a galU mutant of S. sonnei, that is unable to produce a complete LPS with OAg attached, can still assemble OAg material on the cell surface, but a galU mutant of S. flexneri 2a cannot. High molecular weight material not linked to the LPS was purified from S. sonnei and confirmed by NMR to contain the specific sugars of the S. sonnei OAg. Deletion of genes homologous to the group 4 capsule synthesis cluster, previously described in Escherichia coli, abolished the generation of the high molecular weight OAg material. This OAg capsule strongly affects the virulence of S. sonnei. Uncapsulated knockout bacteria were highly invasive in vitro and strongly inflammatory in the rabbit intestine. But, the lack of capsule reduced the ability of S. sonnei to resist complement-mediated killing and to spread from the gut to peripheral organs. In contrast, overexpression of the capsule decreased invasiveness in vitro and inflammation in vivo compared to the wild type. In conclusion, the data indicate that in S. sonnei expression of the capsule modulates bacterial pathogenesis resulting in balanced capabilities to invade and persist in the host environment.


Asunto(s)
Cápsulas Bacterianas/metabolismo , Antígenos O/biosíntesis , Shigella sonnei/metabolismo , Shigella sonnei/patogenicidad , Animales , Cápsulas Bacterianas/genética , Técnicas de Silenciamiento del Gen , Antígenos O/genética , Conejos , Shigella sonnei/genética
16.
PLoS Genet ; 10(10): e1004688, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25356849

RESUMEN

Spinster homolog 2 (Spns2) acts as a Sphingosine-1-phosphate (S1P) transporter in zebrafish and mice, regulating heart development and lymphocyte trafficking respectively. S1P is a biologically active lysophospholipid with multiple roles in signalling. The mechanism of action of Spns2 is still elusive in mammals. Here, we report that Spns2-deficient mice rapidly lost auditory sensitivity and endocochlear potential (EP) from 2 to 3 weeks old. We found progressive degeneration of sensory hair cells in the organ of Corti, but the earliest defect was a decline in the EP, suggesting that dysfunction of the lateral wall was the primary lesion. In the lateral wall of adult mutants, we observed structural changes of marginal cell boundaries and of strial capillaries, and reduced expression of several key proteins involved in the generation of the EP (Kcnj10, Kcnq1, Gjb2 and Gjb6), but these changes were likely to be secondary. Permeability of the boundaries of the stria vascularis and of the strial capillaries appeared normal. We also found focal retinal degeneration and anomalies of retinal capillaries together with anterior eye defects in Spns2 mutant mice. Targeted inactivation of Spns2 in red blood cells, platelets, or lymphatic or vascular endothelial cells did not affect hearing, but targeted ablation of Spns2 in the cochlea using a Sox10-Cre allele produced a similar auditory phenotype to the original mutation, suggesting that local Spns2 expression is critical for hearing in mammals. These findings indicate that Spns2 is required for normal maintenance of the EP and hence for normal auditory function, and support a role for S1P signalling in hearing.


Asunto(s)
Proteínas de Transporte de Anión/genética , Cóclea/patología , Oído Interno/patología , Pérdida Auditiva/genética , Edad de Inicio , Animales , Proteínas de Transporte de Anión/deficiencia , Proteínas de Transporte de Anión/metabolismo , Segmento Anterior del Ojo/metabolismo , Segmento Anterior del Ojo/patología , Cóclea/metabolismo , Conexina 26 , Conexinas , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Pérdida Auditiva/metabolismo , Pérdida Auditiva/patología , Lisofosfolípidos/metabolismo , Ratones , Organogénesis/genética , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Estría Vascular/patología , Pez Cebra
17.
Infect Immun ; 83(7): 2926-34, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25964470

RESUMEN

The intestinal mucosa forms the first line of defense against infections mediated by enteric pathogens such as salmonellae. Here we exploited intestinal "organoids" (iHOs) generated from human induced pluripotent stem cells (hIPSCs) to explore the interaction of Salmonella enterica serovar Typhimurium with iHOs. Imaging and RNA sequencing were used to analyze these interactions, and clear changes in transcriptional signatures were detected, including altered patterns of cytokine expression after the exposure of iHOs to bacteria. S. Typhimurium microinjected into the lumen of iHOs was able to invade the epithelial barrier, with many bacteria residing within Salmonella-containing vacuoles. An S. Typhimurium invA mutant defective in the Salmonella pathogenicity island 1 invasion apparatus was less capable of invading the iHO epithelium. Hence, we provide evidence that hIPSC-derived organoids are a promising model of the intestinal epithelium for assessing interactions with enteric pathogens.


Asunto(s)
Interacciones Huésped-Patógeno , Células Madre Pluripotentes Inducidas/microbiología , Células Madre Pluripotentes Inducidas/fisiología , Organoides/microbiología , Organoides/fisiología , Salmonella typhimurium/crecimiento & desarrollo , Proteínas Bacterianas/genética , Citocinas/metabolismo , Células Epiteliales/microbiología , Eliminación de Gen , Perfilación de la Expresión Génica , Humanos , Modelos Teóricos , Imagen Óptica , Vacuolas/microbiología
18.
PLoS Pathog ; 9(12): e1003842, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24367267

RESUMEN

Group A streptococcal isolates of serotype M18 are historically associated with epidemic waves of pharyngitis and the non-suppurative immune sequela rheumatic fever. The serotype is defined by a unique, highly encapsulated phenotype, yet the molecular basis for this unusual colony morphology is unknown. Here we identify a truncation in the regulatory protein RocA, unique to and conserved within our serotype M18 GAS collection, and demonstrate that it underlies the characteristic M18 capsule phenotype. Reciprocal allelic exchange mutagenesis of rocA between M18 GAS and M89 GAS demonstrated that truncation of RocA was both necessary and sufficient for hyper-encapsulation via up-regulation of both precursors required for hyaluronic acid synthesis. Although RocA was shown to positively enhance covR transcription, quantitative proteomics revealed RocA to be a metabolic regulator with activity beyond the CovR/S regulon. M18 GAS demonstrated a uniquely protuberant chain formation following culture on agar that was dependent on excess capsule and the RocA mutation. Correction of the M18 rocA mutation reduced GAS survival in human blood, and in vivo naso-pharyngeal carriage longevity in a murine model, with an associated drop in bacterial airborne transmission during infection. In summary, a naturally occurring truncation in a regulator explains the encapsulation phenotype, carriage longevity and transmissibility of M18 GAS, highlighting the close interrelation of metabolism, capsule and virulence.


Asunto(s)
Viabilidad Microbiana/genética , Streptococcus/fisiología , Transactivadores/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células Cultivadas , Codón sin Sentido , Femenino , Humanos , Ratones , Datos de Secuencia Molecular , Nasofaringe/microbiología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/transmisión , Serotipificación , Esporas Bacterianas/genética , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/transmisión , Streptococcus/clasificación , Streptococcus/crecimiento & desarrollo , Streptococcus pyogenes/crecimiento & desarrollo , Streptococcus pyogenes/fisiología
19.
Proc Natl Acad Sci U S A ; 109(6): 2108-13, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22308390

RESUMEN

Bifidobacteria comprise a significant proportion of the human gut microbiota. Several bifidobacterial strains are currently used as therapeutic interventions, claiming various health benefits by acting as probiotics. However, the precise mechanisms by which they maintain habitation within their host and consequently provide these benefits are not fully understood. Here we show that Bifidobacterium breve UCC2003 produces a cell surface-associated exopolysaccharide (EPS), the biosynthesis of which is directed by either half of a bidirectional gene cluster, thus leading to production of one of two possible EPSs. Alternate transcription of the two opposing halves of this cluster appears to be the result of promoter reorientation. Surface EPS provided stress tolerance and promoted in vivo persistence, but not initial colonization. Marked differences were observed in host immune response: strains producing surface EPS (EPS(+)) failed to elicit a strong immune response compared with EPS-deficient variants. Specifically, EPS production was shown to be linked to the evasion of adaptive B-cell responses. Furthermore, presence of EPS(+) B. breve reduced colonization levels of the gut pathogen Citrobacter rodentium. Our data thus assigns a pivotal and beneficial role for EPS in modulating various aspects of bifidobacterial-host interaction, including the ability of commensal bacteria to remain immunologically silent and in turn provide pathogen protection. This finding enforces the probiotic concept and provides mechanistic insights into health-promoting benefits for both animal and human hosts.


Asunto(s)
Bifidobacterium/inmunología , Membrana Celular/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad/inmunología , Polisacáridos Bacterianos/inmunología , Ácidos , Animales , Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Infecciones por Bifidobacteriales/inmunología , Infecciones por Bifidobacteriales/microbiología , Bifidobacterium/crecimiento & desarrollo , Bilis , Citrobacter/crecimiento & desarrollo , Recuento de Colonia Microbiana , Citocinas/metabolismo , Sistema Digestivo/microbiología , Sitios Genéticos/genética , Humanos , Evasión Inmune/inmunología , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Bazo/inmunología , Bazo/microbiología
20.
BMC Genomics ; 15: 160, 2014 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-24568651

RESUMEN

BACKGROUND: Clostridium difficile is an anaerobic, Gram-positive bacterium that can reside as a commensal within the intestinal microbiota of healthy individuals or cause life-threatening antibiotic-associated diarrhea in immunocompromised hosts. C. difficile can also form highly resistant spores that are excreted facilitating host-to-host transmission. The C. difficile spo0A gene encodes a highly conserved transcriptional regulator of sporulation that is required for relapsing disease and transmission in mice. RESULTS: Here we describe a genome-wide approach using a combined transcriptomic and proteomic analysis to identify Spo0A regulated genes. Our results validate Spo0A as a positive regulator of putative and novel sporulation genes as well as components of the mature spore proteome. We also show that Spo0A regulates a number of virulence-associated factors such as flagella and metabolic pathways including glucose fermentation leading to butyrate production. CONCLUSIONS: The C. difficile spo0A gene is a global transcriptional regulator that controls diverse sporulation, virulence and metabolic phenotypes coordinating pathogen adaptation to a wide range of host interactions. Additionally, the rich breadth of functional data allowed us to significantly update the annotation of the C. difficile 630 reference genome which will facilitate basic and applied research on this emerging pathogen.


Asunto(s)
Clostridioides difficile/fisiología , Clostridioides difficile/patogenicidad , Redes y Vías Metabólicas , Proteoma , Transcriptoma , Butiratos/metabolismo , Clostridioides difficile/crecimiento & desarrollo , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Glucosa/metabolismo , Humanos , Anotación de Secuencia Molecular , Datos de Secuencia Molecular , Mutación , Proteómica , Esporas Bacterianas , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA