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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 18(3): e1009776, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35286304

RESUMEN

Shotgun metagenomics is a powerful tool to identify antimicrobial resistance (AMR) genes in microbiomes but has the limitation that extrachromosomal DNA, such as plasmids, cannot be linked with the host bacterial chromosome. Here we present a comprehensive laboratory and bioinformatics pipeline HAM-ART (Hi-C Assisted Metagenomics for Antimicrobial Resistance Tracking) optimised for the generation of metagenome-assembled genomes including both chromosomal and extrachromosomal AMR genes. We demonstrate the performance of the pipeline in a study comparing 100 pig faecal microbiomes from low- and high-antimicrobial use pig farms (organic and conventional farms). We found significant differences in the distribution of AMR genes between low- and high-antimicrobial use farms including a plasmid-borne lincosamide resistance gene exclusive to high-antimicrobial use farms in three species of Lactobacilli. The bioinformatics pipeline code is available at https://github.com/lkalmar/HAM-ART.


Asunto(s)
Antiinfecciosos , Microbiota , Animales , Antibacterianos , Antiinfecciosos/farmacología , Farmacorresistencia Bacteriana/genética , Metagenómica , Porcinos
2.
BMC Biol ; 20(1): 14, 2022 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-35027054

RESUMEN

BACKGROUND: Infectious diseases of farmed and wild animals pose a recurrent threat to food security and human health. The macrophage, a key component of the innate immune system, is the first line of defence against many infectious agents and plays a major role in shaping the adaptive immune response. However, this phagocyte is a target and host for many pathogens. Understanding the molecular basis of interactions between macrophages and pathogens is therefore crucial for the development of effective strategies to combat important infectious diseases. RESULTS: We explored how porcine pluripotent stem cells (PSCs) can provide a limitless in vitro supply of genetically and experimentally tractable macrophages. Porcine PSC-derived macrophages (PSCdMs) exhibited molecular and functional characteristics of ex vivo primary macrophages and were productively infected by pig pathogens, including porcine reproductive and respiratory syndrome virus (PRRSV) and African swine fever virus (ASFV), two of the most economically important and devastating viruses in pig farming. Moreover, porcine PSCdMs were readily amenable to genetic modification by CRISPR/Cas9 gene editing applied either in parental stem cells or directly in the macrophages by lentiviral vector transduction. CONCLUSIONS: We show that porcine PSCdMs exhibit key macrophage characteristics, including infection by a range of commercially relevant pig pathogens. In addition, genetic engineering of PSCs and PSCdMs affords new opportunities for functional analysis of macrophage biology in an important livestock species. PSCs and differentiated derivatives should therefore represent a useful and ethical experimental platform to investigate the genetic and molecular basis of host-pathogen interactions in pigs, and also have wider applications in livestock.


Asunto(s)
Virus de la Fiebre Porcina Africana , Enfermedades Transmisibles , Virus de la Fiebre Porcina Africana/genética , Animales , Interacciones Huésped-Patógeno/genética , Macrófagos , Células Madre , Porcinos
3.
Immunology ; 165(2): 171-194, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34767637

RESUMEN

Conventional dendritic cells (cDC) are bone marrow-derived immune cells that play a central role in linking innate and adaptive immunity. cDCs efficiently uptake, process and present antigen to naïve T cells, driving clonal expansion of antigen-specific T-cell responses. In chicken, vital reagents are lacking for the efficient and precise identification of cDCs. In this study, we have developed several novel reagents for the identification and characterization of chicken cDCs. Chicken FLT3 cDNA was cloned and a monoclonal antibody to cell surface FLT3 was generated. This antibody identified a distinct FLT3HI splenic subset which lack expression of signature markers for B cells, T cells or monocyte/macrophages. By combining anti-FLT3 and CSF1R-eGFP transgenic expression, three major populations within the mononuclear phagocyte system were identified in the spleen. The cDC1 subset of mammalian cDCs express the chemokine receptor XCR1. To characterize chicken cDCs, a synthetic chicken chemokine (C motif) ligand (XCL1) peptide conjugated to Alexa Fluor 647 was developed (XCL1AF647 ). Flow cytometry staining of XCL1AF647 on splenocytes showed that all chicken FLT3HI cells exclusively express XCR1, supporting the hypothesis that this population comprises bona fide chicken cDCs. Further analysis revealed that chicken cDCs expressed CSF1R but lacked the expression of CSF2R. Collectively, the cell surface phenotypes of chicken cDCs were partially conserved with mammalian XCR1+ cDC1, with distinct differences in CSF1R and CSF2R expression compared with mammalian orthologues. These original reagents allow the efficient identification of chicken cDCs to investigate their important roles in the chicken immunity and diseases.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Anticuerpos Monoclonales , Biomarcadores , Técnicas de Cultivo de Célula , Pollos , Técnica del Anticuerpo Fluorescente , Expresión Génica , Humanos , Inmunofenotipificación , Receptores Acoplados a Proteínas G/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Tirosina Quinasa 3 Similar a fms/genética
4.
Microb Cell Fact ; 21(1): 181, 2022 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-36071433

RESUMEN

BACKGROUND: Worldwide, an estimated 70.7 billion broilers were produced in 2020. With the reduction in use of prophylactic antibiotics as a result of consumer pressure and regulatory oversight alternative approaches, such as vaccination, are required to control bacterial infections. A potential way to produce a multivalent vaccine is via the generation of a glycoconjugate vaccine which consists of an antigenic protein covalently linked to an immunogenic carbohydrate. Protein-glycan coupling technology (PGCT) is an approach to generate glycoconjugates using enzymes that can couple proteins and glycan when produced in bacterial cells. Previous studies have used PGCT to generate a live-attenuated avian pathogenic Escherichia coli (APEC) strain capable of N-glycosylation of target proteins using a chromosomally integrated Campylobacter jejuni pgl locus. However, this proved ineffective against C. jejuni challenge. RESULTS: In this study we demonstrate the lack of surface exposure of glycosylated protein in APEC strain χ7122 carrying the pgl locus. Furthermore, we hypothesise that this may be due to the complex cell-surface architecture of E. coli. To this end, we removed the lipopolysaccharide O-antigen of APEC χ7122 pgl+ via deletion of the wecA gene and demonstrate increased surface exposure of glycosylated antigens (NetB and FlpA) in this strain. We hypothesise that increasing the surface expression of the glycosylated protein would increase the chance of host immune cells being exposed to the glycoconjugate, and therefore the generation of an efficacious immune response would be more likely. CONCLUSIONS: Our results demonstrate an increase in cell surface exposure and therefore accessibility of glycosylated antigens upon removal of lipopolysaccharide antigen from the APEC cell surface.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli , Animales , Pollos , Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/prevención & control , Infecciones por Escherichia coli/veterinaria , Glicoconjugados , Lipopolisacáridos
5.
Microb Cell Fact ; 21(1): 6, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34986868

RESUMEN

BACKGROUND: Campylobacter is an animal and zoonotic pathogen of global importance, and a pressing need exists for effective vaccines, including those that make use of conserved polysaccharide antigens. To this end, we adapted Protein Glycan Coupling Technology (PGCT) to develop a versatile Escherichia coli strain capable of generating multiple glycoconjugate vaccine candidates against Campylobacter jejuni. RESULTS: We generated a glycoengineering E. coli strain containing the conserved C. jejuni heptasaccharide coding region integrated in its chromosome as a model glycan. This methodology confers three advantages: (i) reduction of plasmids and antibiotic markers used for PGCT, (ii) swift generation of many glycan-protein combinations and consequent rapid identification of the most antigenic proteins or peptides, and (iii) increased genetic stability of the polysaccharide coding-region. In this study, by using the model glycan expressing strain, we were able to test proteins from C. jejuni, Pseudomonas aeruginosa (both Gram-negative), and Clostridium perfringens (Gram-positive) as acceptors. Using this pgl integrant E. coli strain, four glycoconjugates were readily generated. Two glycoconjugates, where both protein and glycan are from C. jejuni (double-hit vaccines), and two glycoconjugates, where the glycan antigen is conjugated to a detoxified toxin from a different pathogen (single-hit vaccines). Because the downstream application of Live Attenuated Vaccine Strains (LAVS) against C. jejuni is to be used in poultry, which have a higher body temperature of 42 °C, we investigated the effect of temperature on protein expression and glycosylation in the E. coli pgl integrant strain. CONCLUSIONS: We determined that glycosylation is temperature dependent and that for the combination of heptasaccharide and carriers used in this study, the level of PglB available for glycosylation is a step limiting factor in the glycosylation reaction. We also demonstrated that temperature affects the ability of PglB to glycosylate its substrates in an in vitro glycosylation assay independent of its transcriptional level.


Asunto(s)
Proteínas Bacterianas/metabolismo , Cromosomas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Glicoconjugados/metabolismo , Temperatura , Proteínas Bacterianas/genética , Vacunas Bacterianas , Campylobacter jejuni/genética , Campylobacter jejuni/inmunología , Glicosilación , Proteínas de la Membrana/genética , Ingeniería Metabólica/métodos , Polisacáridos Bacterianos/genética
6.
BMC Genomics ; 22(1): 411, 2021 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-34082718

RESUMEN

BACKGROUND: Campylobacter jejuni is the leading cause of bacterial gastroenteritis in humans and the handling or consumption of contaminated poultry meat is a key source of infection. Selective breeding of poultry that exhibit elevated resistance to Campylobacter is an attractive control strategy. Here we studied the global transcriptional response of inbred chicken lines that differ in resistance to C. jejuni colonisation at a key site of bacterial persistence. RESULTS: Three-week-old chickens of line 61 and N were inoculated orally with C. jejuni strain M1 and caecal contents and tonsils were sampled at 1 and 5 days post-infection. Caecal colonisation was significantly lower in line 61 compared to line N at 1 day post-infection, but not 5 days post-infection. RNA-Seq analysis of caecal tonsils of both lines revealed a limited response to C. jejuni infection compared to age-matched uninfected controls. In line N at days 1 and 5 post-infection, just 8 and 3 differentially expressed genes (DEGs) were detected (fold-change > 2 and false-discovery rate of < 0.05) relative to uninfected controls, respectively. In the relatively resistant line 61, a broader response to C. jejuni was observed, with 69 DEGs relating to immune regulation, cell signalling and metabolism at 1 day post-infection. However, by day 5 post-infection, no DEGs were detected. By far, the greatest number of DEGs were between uninfected birds of the two lines implying that differential resistance to C. jejuni is intrinsic. Of these genes, several Major Histocompatibility Complex class I-related genes (MHCIA1, MHCBL2 and MHCIY) and antimicrobial peptides (MUC2, AvBD10 and GZMA) were expressed to a greater extent in line N. Two genes within quantitative trait loci associated with C. jejuni colonisation were also more highly expressed in line N (ASIC4 and BZFP2). Quantitative reverse-transcriptase PCR analysis of a subset of transcripts confirmed the RNA-Seq results. CONCLUSIONS: Our data indicate a limited transcriptional response in the caecal tonsils of inbred chickens to intestinal colonisation by Campylobacter but identify a large number of differentially transcribed genes between lines 61 and N that may underlie variation in heritable resistance to C. jejuni.


Asunto(s)
Infecciones por Campylobacter , Campylobacter jejuni , Enfermedades de las Aves de Corral , Animales , Infecciones por Campylobacter/genética , Infecciones por Campylobacter/veterinaria , Campylobacter jejuni/genética , Ciego , Pollos/genética , Perfilación de la Expresión Génica , Humanos , Enfermedades de las Aves de Corral/genética , Transcriptoma
7.
Microb Cell Fact ; 20(1): 193, 2021 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-34600535

RESUMEN

BACKGROUND: Poultry is the world's most popular animal-based food and global production has tripled in the past 20 years alone. Low-cost vaccines that can be combined to protect poultry against multiple infections are a current global imperative. Glycoconjugate vaccines, which consist of an immunogenic protein covalently coupled to glycan antigens of the targeted pathogen, have a proven track record in human vaccinology, but have yet to be used for livestock due to prohibitively high manufacturing costs. To overcome this, we use Protein Glycan Coupling Technology (PGCT), which enables the production of glycoconjugates in bacterial cells at considerably reduced costs, to generate a candidate glycan-based live vaccine intended to simultaneously protect against Campylobacter jejuni, avian pathogenic Escherichia coli (APEC) and Clostridium perfringens. Campylobacter is the most common cause of food poisoning, whereas colibacillosis and necrotic enteritis are widespread and devastating infectious diseases in poultry. RESULTS: We demonstrate the functional transfer of C. jejuni protein glycosylation (pgl) locus into the genome of APEC χ7122 serotype O78:H9. The integration caused mild attenuation of the χ7122 strain following oral inoculation of chickens without impairing its ability to colonise the respiratory tract. We exploit the χ7122 pgl integrant as bacterial vectors delivering a glycoprotein decorated with the C. jejuni heptasaccharide glycan antigen. To this end we engineered χ7122 pgl to express glycosylated NetB toxoid from C. perfringens and tested its ability to reduce caecal colonisation of chickens by C. jejuni and protect against intra-air sac challenge with the homologous APEC strain. CONCLUSIONS: We generated a candidate glycan-based multivalent live vaccine with the potential to induce protection against key avian and zoonotic pathogens (C. jejuni, APEC, C. perfringens). The live vaccine failed to significantly reduce Campylobacter colonisation under the conditions tested but was protective against homologous APEC challenge. Nevertheless, we present a strategy towards the production of low-cost "live-attenuated multivalent vaccine factories" with the ability to express glycoconjugates in poultry.


Asunto(s)
Infecciones por Campylobacter/prevención & control , Infecciones por Clostridium/prevención & control , Infecciones por Escherichia coli/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Desarrollo de Vacunas/métodos , Animales , Campylobacter jejuni/inmunología , Pollos , Clostridium perfringens/inmunología , Escherichia coli/inmunología , Vacunas Atenuadas/inmunología , Vacunas Combinadas/inmunología
8.
Avian Pathol ; 50(5): 382-391, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33410704

RESUMEN

Avian pathogenic Escherichia coli (APEC) cause colibacillosis in birds, a syndrome of severe respiratory and systemic disease that constitutes a major threat due to early mortality, condemnation of carcasses and reduced productivity. APEC can infect different types of birds in all commercial settings, and birds of all ages, although disease tends to be more severe in younger birds likely a consequence of an immature immune system. APEC can act as both primary and secondary pathogens, with predisposing factors for secondary infections including poor housing conditions, respiratory viral and Mycoplasma spp. infections or vaccinations. Controlled studies with APEC as primary pathogens have been used to study the bird's immune response to APEC, although it may not always be representative of natural infections which may be more complex due to the presence of secondary agents, stress and environmental factors. Under controlled experimental conditions, a strong early innate immune response is induced which includes host defence peptides in mucus and a cellular response driven by heterophils and macrophages. Both antibody and T-cell mediated adaptive responses have been demonstrated after vaccination. In this review we will discuss the bird's immune response to APEC as primary pathogen with a bias towards the innate immune response, as mechanistic adaptive studies clearly form a much more limited body of work despite numerous vaccine trials.


Asunto(s)
Aves/inmunología , Escherichia coli , Animales , Aves/microbiología , Calidad de la Vivienda , Inmunidad
9.
Appl Environ Microbiol ; 86(7)2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-31980428

RESUMEN

Campylobacteriosis is the leading foodborne bacterial diarrheal illness in many countries, with up to 80% of human cases attributed to the avian reservoir. The only control strategies currently available are stringent on-farm biosecurity and carcass treatments. Heritable differences in the resistance of chicken lines to Campylobacter colonization have been reported and resistance-associated quantitative trait loci are emerging, although their impact on colonization appears modest. Recent studies indicated a protective role of the microbiota against colonization by Campylobacter in chickens. Furthermore, in murine models, differences in resistance to bacterial infections can be partially transferred between lines by transplantation of gut microbiota. In this study, we investigated whether heritable differences in colonization of inbred chicken lines by Campylobacter jejuni are associated with differences in cecal microbiota. We performed homologous and heterologous cecal microbiota transplants between line 61 (resistant) and line N (susceptible) by orally administering cecal contents collected from 3-week-old donors to day-of-hatch chicks. Recipient birds were challenged (day 21) with C. jejuni 11168H. In birds given homologous microbiota, the differential resistance of lines to C. jejuni colonization was reproduced. Contrary to our hypothesis, transfer of cecal microbiota from line 61 to line N significantly increased C. jejuni colonization. No significant difference in the overall composition of the cecal microbial communities of the two lines was identified, although line-specific differences for specific operational taxonomic units were identified. Our data suggest that while heritable differences in avian resistance to Campylobacter colonization exist, these are not explained by significant variation in the cecal microbiota.IMPORTANCECampylobacter is a leading cause of foodborne diarrheal disease worldwide. Poultry are a key source of human infections, but there are currently few effective measures against Campylobacter in poultry during production. One option to control Campylobacter may be to alter the composition of microbial communities in the avian intestines by introducing beneficial bacteria, which exclude the harmful ones. We previously described two inbred chicken lines which differ in resistance to intestinal colonization by Campylobacter Here, we investigated the composition of the microbial communities in the gut of these lines and whether transferring gut bacteria between the resistant and susceptible lines alters their resistance to Campylobacter No major differences in microbial populations were found, and resistance or susceptibility to colonization was not conferred by transferring gut bacteria between lines. The data suggest that gut microbiota did not play a role in resistance to Campylobacter colonization, at least in the lines used.


Asunto(s)
Infecciones por Campylobacter/veterinaria , Campylobacter jejuni/fisiología , Ciego/microbiología , Pollos , Resistencia a la Enfermedad , Microbioma Gastrointestinal , Enfermedades de las Aves de Corral/microbiología , Animales , Infecciones por Campylobacter/microbiología , Pollos/genética , Femenino , Endogamia , Masculino
10.
Infect Immun ; 87(2)2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30510107

RESUMEN

Eimeria tenella can cause the disease coccidiosis in chickens. The direct and often detrimental impact of this parasite on chicken health, welfare, and productivity is well recognized; however, less is known about the secondary effects that infection may have on other gut pathogens. Campylobacter jejuni is the leading cause of human bacterial foodborne disease in many countries and has been demonstrated to exert negative effects on poultry welfare and production in some broiler lines. Previous studies have shown that concurrent Eimeria infection can influence the colonization and replication of bacteria, such as Clostridium perfringens and Salmonella enterica serovar Typhimurium. Through a series of in vivo coinfection experiments, this study evaluated the impact that E. tenella infection had on C. jejuni colonization of chickens, including the influence of variations in parasite dose and sampling time after bacterial challenge. Coinfection with E. tenella resulted in a significant increase in C. jejuni colonization in the cecum in a parasite dose-dependent manner but a significant decrease in C. jejuni colonization in the spleen and liver of chickens. The results were reproducible at 3 and 10 days after bacterial infection. This work highlights that E. tenella not only has a direct impact on the health and well-being of chickens but can have secondary effects on important zoonotic pathogens.


Asunto(s)
Infecciones por Campylobacter/microbiología , Campylobacter jejuni/aislamiento & purificación , Pollos/microbiología , Coccidiosis/complicaciones , Coinfección , Eimeria tenella , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/parasitología , Animales , Ciego/microbiología , Coinfección/microbiología , Coinfección/parasitología
11.
BMC Genomics ; 20(1): 20, 2019 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-30621582

RESUMEN

BACKGROUND: Salmonella enterica subspecies enterica is an animal and zoonotic pathogen of global importance. Cattle are a significant reservoir of human non-typhoidal salmonellosis and can suffer enteric and systemic disease owing to the ability of Salmonella to survive within the bovine lymphatic system and intestines. Contamination of food can occur due to the incorporation of contaminated peripheral lymph nodes or by direct contamination of carcasses with gut contents. It is essential to understand the mechanisms used by Salmonella to enter and persist within the bovine lymphatic system and how they differ from those required for intestinal colonization to minimize zoonotic infections. RESULTS: Transposon-directed insertion site sequencing (TraDIS) was applied to pools of mutants recovered from mesenteric lymph nodes (MLNs) draining the distal ileum of calves after oral inoculation with a library of 8550 random S. Typhimurium mini-Tn5Km2 mutants in pools of 475 mutants per calf. A total of 8315 mutants representing 2852 different genes were detected in MLNs and their in vivo fitness was calculated. Using the same improved algorithm for analysis of transposon-flanking sequences, the identity and phenotype of mutants recovered from the distal ileal mucosa of the same calves was also defined, enabling comparison with previously published data and of mutant phenotypes across the tissues. Phenotypes observed for the majority of mutants were highly significantly correlated in the two tissues. However, 32 genes were identified in which transposon insertions consistently resulted in differential fitness in the ileal wall and MLNs, suggesting niche-specific roles for these genes in pathogenesis. Defined null mutations affecting ptsN and spvC were confirmed to result in tissue-specific phenotypes in calves, thus validating the TraDIS dataset. CONCLUSIONS: This validation of the role of thousands of Salmonella genes and identification of genes with niche-specific roles in a key target species will inform the design of control strategies for bovine salmonellosis and zoonotic infections, for which efficacious and cross-protective vaccines are currently lacking.


Asunto(s)
Elementos Transponibles de ADN/genética , Infecciones por Salmonella/genética , Salmonella enterica/genética , Salmonella typhimurium/genética , Animales , Liasas de Carbono-Oxígeno/genética , Bovinos , Enfermedades de los Bovinos/genética , Enfermedades de los Bovinos/microbiología , Reservorios de Enfermedades/microbiología , Humanos , Íleon/microbiología , Intestinos/microbiología , Ganglios Linfáticos/microbiología , Mutación , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/transmisión , Salmonella enterica/patogenicidad , Salmonella typhimurium/patogenicidad
12.
Vet Res ; 50(1): 99, 2019 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-31771636

RESUMEN

Salmonella enterica is a veterinary and zoonotic pathogen of global importance. While murine and cell-based models of infection have provided considerable knowledge about the molecular basis of virulence of Salmonella, relatively little is known about salmonellosis in naturally-affected large animal hosts such as cattle, which are a reservoir of human salmonellosis. As in humans, Salmonella causes bovine disease ranging from self-limiting enteritis to systemic typhoid-like disease and exerts significant economic and welfare costs. Understanding the nature and consequences of Salmonella interactions with bovine cells will inform the design of effective vaccines and interventions to control animal and zoonotic infections. In calves challenged orally with S. Dublin expressing green fluorescent protein (GFP) we observed that the bacteria were predominantly extracellular in the distal ileal mucosa and within gut-associated lymph nodes 48 h post-infection. Intracellular bacteria, identified by flow cytometry using the GFP signal, were predominantly within MHCII+ macrophage-like cells. In contrast to observations from murine models, these S. Dublin-infected cells had elevated levels of MHCII and CD40 compared to both uninfected cells from the same tissue and cells from the cognate tissue of uninfected animals. Moreover, no gross changes of the architecture of infected lymph nodes were observed as was described previously in a mouse model. In order to further investigate Salmonella-macrophage interactions, net replication of S. enterica serovars that differ in virulence in cattle was measured in bovine blood-derived macrophages by enumeration of gentamicin-protected bacteria and fluorescence dilution, but did not correlate with host-specificity.


Asunto(s)
Enfermedades de los Bovinos/fisiopatología , Salmonelosis Animal/fisiopatología , Salmonella enterica/fisiología , Animales , Bovinos , Enfermedades de los Bovinos/microbiología , Antígenos de Histocompatibilidad Clase II/análisis , Salmonelosis Animal/microbiología , Serogrupo
13.
Avian Pathol ; 48(2): 157-167, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30570345

RESUMEN

Avian pathogenic E. coli (APEC) cause severe respiratory and systemic disease. To address the genetic and immunological basis of resistance, inbred chicken lines were used to establish a model of differential resistance to APEC, using strain O1 of serotype O1:K1:H7. Inbred lines 72, 15I and C.B12 and the outbred line Novogen Brown were inoculated via the airsac with a high dose (107 colony-forming units, CFU) or low dose (105 CFU) of APEC O1. Clinical signs, colibacillosis lesion score and bacterial colonization of tissues after high dose challenge were significantly higher in line 15I and C.B12 birds. The majority of the 15I and C.B12 birds succumbed to the infection by 14 h post-infection, whilst none of the line 72 and the Novogen Brown birds developed clinical signs. No difference was observed after low dose challenge. In a repeat study, inbred lines 72 and 15I were inoculated with low, intermediate or high doses of APEC O1 ranging from 105 to 107 CFU. The colonization of lung was highest in line 15I after high dose challenge and birds developed clinical signs; however, colonization of blood and spleen, clinical signs and lesion score were not different between lines. No difference was observed after intermediate or low dose challenge. Ex vivo, the phagocytic and bactericidal activity of lung leukocytes from line 72 and 15I birds did not differ. Our data suggest that although differential resistance of inbred lines 72, 15I and C.B12 to APEC O1 challenge is apparent, it is dependent on the infectious dose. Research Highlights Lines 15I and C.B12 are more susceptible than line 72 to a high dose of APEC O1. Differential resistance is dose-dependent in lines 15I and 72. Phagocytic and bactericidal activity is similar and dose independent.


Asunto(s)
Pollos , Resistencia a la Enfermedad , Infecciones por Escherichia coli/veterinaria , Escherichia coli/inmunología , Inmunidad Innata , Enfermedades de las Aves de Corral/inmunología , Sacos Aéreos/microbiología , Animales , Animales Endogámicos , Anticuerpos Heterófilos/inmunología , Carga Bacteriana , Relación Dosis-Respuesta Inmunológica , Escherichia coli/genética , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Femenino , Macrófagos/inmunología , Masculino , Enfermedades de las Aves de Corral/microbiología , Organismos Libres de Patógenos Específicos
14.
Infect Immun ; 86(5)2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29339462

RESUMEN

Gastrointestinal bacterial infection represents a significant threat to human health, as well as a burden on food animal production and welfare. Although there is advanced knowledge about the molecular mechanisms underlying pathogenesis, including the development of immune responses to these pathogens, gaps in knowledge persist. It is well established that gastrointestinal bacterial pathogens produce a myriad of proteins that affect the development and effectiveness of innate immune responses. However, relatively few proteins that directly affect lymphocytes responsible for humoral or cell-mediated immunity and memory have been identified. Here, we review factors produced by gastrointestinal bacterial pathogens that have direct T cell interactions and what is known about their functions and mechanisms of action. T cell-interacting bacterial proteins that have been identified to date mainly target three major T cell responses: activation and expansion, chemotaxis, or apoptosis. Further, the requirement for more focused studies to identify and understand additional mechanisms used by bacteria to directly affect the T cell immune response and how these may contribute to pathogenesis is highlighted. Increased knowledge in this area will help to drive development of better interventions in prevention and treatment of gastrointestinal bacterial infection.


Asunto(s)
Apoptosis/inmunología , Enfermedades Gastrointestinales/inmunología , Enfermedades Gastrointestinales/microbiología , Microbioma Gastrointestinal/inmunología , Inmunidad Innata/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Humanos
15.
BMC Genomics ; 19(1): 594, 2018 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-30086717

RESUMEN

BACKGROUND: The domestic chicken (Gallus gallus) is widely used as a model in developmental biology and is also an important livestock species. We describe a novel approach to data integration to generate an mRNA expression atlas for the chicken spanning major tissue types and developmental stages, using a diverse range of publicly-archived RNA-seq datasets and new data derived from immune cells and tissues. RESULTS: Randomly down-sampling RNA-seq datasets to a common depth and quantifying expression against a reference transcriptome using the mRNA quantitation tool Kallisto ensured that disparate datasets explored comparable transcriptomic space. The network analysis tool Graphia was used to extract clusters of co-expressed genes from the resulting expression atlas, many of which were tissue or cell-type restricted, contained transcription factors that have previously been implicated in their regulation, or were otherwise associated with biological processes, such as the cell cycle. The atlas provides a resource for the functional annotation of genes that currently have only a locus ID. We cross-referenced the RNA-seq atlas to a publicly available embryonic Cap Analysis of Gene Expression (CAGE) dataset to infer the developmental time course of organ systems, and to identify a signature of the expansion of tissue macrophage populations during development. CONCLUSION: Expression profiles obtained from public RNA-seq datasets - despite being generated by different laboratories using different methodologies - can be made comparable to each other. This meta-analytic approach to RNA-seq can be extended with new datasets from novel tissues, and is applicable to any species.


Asunto(s)
Pollos/genética , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia de ARN/métodos , Animales , Atlas como Asunto , Bases de Datos Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento
16.
Appl Environ Microbiol ; 84(4)2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29180370

RESUMEN

Salmonella enterica is an animal and zoonotic pathogen of worldwide importance. Salmonella serovars that differ in their host and tissue tropisms exist. Cattle are an important reservoir of human nontyphoidal salmonellosis, and contaminated bovine peripheral lymph nodes enter the food chain via ground beef. The relative abilities of different serovars to survive within the bovine lymphatic system are poorly understood and constrain the development of control strategies. This problem was addressed by developing a massively parallel whole-genome sequencing method to study mixed-serovar infections in vivoSalmonella serovars differ genetically by naturally occurring single nucleotide polymorphisms (SNPs) in certain genes. It was hypothesized that these SNPs could be used as markers to simultaneously identify serovars in mixed populations and quantify the abundance of each member in a population. The performance of the method was validated in vitro using simulated pools containing up to 11 serovars in various proportions. It was then applied to study serovar survival in vivo in cattle challenged orally with the same 11 serovars. All the serovars successfully colonized the bovine lymphatic system, including the peripheral lymph nodes, and thus pose similar risks of zoonosis. This method enables the fates of multiple genetically unmodified strains to be evaluated simultaneously in a single animal. It could be useful in reducing the number of animals required to study mixed-strain infections and in testing the cross-protective efficacy of vaccines and treatments. It also has the potential to be applied to diverse bacterial species which possess shared but polymorphic alleles.IMPORTANCE While some Salmonella serovars are more frequently isolated from lymph nodes rather than the feces and environment of cattle, the relative abilities of serovars to survive within the lymphatic system of cattle remain ill defined. A sequencing-based method which used available information from sequenced Salmonella genomes to study the dynamics of mixed-serovar infections in vivo was developed. The main advantages of the method include the simultaneous identification and quantification of multiple strains without any genetic modification and minimal animal use. This approach could be used in vaccination trials or in epidemiological surveys where an understanding of the dynamics of closely related strains of a pathogen in mixed populations could inform the prediction of zoonotic risk and the development of intervention strategies.


Asunto(s)
Enfermedades de los Bovinos/epidemiología , Polimorfismo de Nucleótido Simple , Salmonelosis Animal/epidemiología , Salmonella enterica/fisiología , Secuenciación Completa del Genoma/veterinaria , Animales , Bovinos , Enfermedades de los Bovinos/microbiología , Longevidad , Factores de Riesgo , Salmonelosis Animal/microbiología , Salmonella enterica/genética , Serogrupo , Zoonosis/epidemiología , Zoonosis/microbiología
17.
Vet Res ; 49(1): 3, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29316981

RESUMEN

Campylobacter jejuni is the leading cause of bacterial food-borne gastroenteritis worldwide and human infections are frequently associated with handling and consumption of contaminated poultry. The polysaccharide capsule of C. jejuni plays important roles in colonisation of the chicken gut, invasion of epithelial cells and serum resistance and is subject to modification with O-methyl phosphoramidate (MeOPN) in most strains. In this study, the cytokine responses of mouse bone marrow-derived macrophages (mBMMs), chicken bone marrow-derived macrophages (chBMMs) and human monocyte-derived macrophages (hMDMs) were measured following infection with C. jejuni 11168H wild-type (WT) or isogenic mutants lacking either the capsule (Δcj1439) or its MeOPN modification (Δcj1417). Consistent with previous observations using murine bone marrow-derived dendritic cells, mutants lacking the capsule or MeOPN elicited enhanced transcription of IL-6 and IL-10 in mBMMs compared to wild-type C. jejuni. However, the lack of capsule and MeOPN did not alter IL-6 and IL-10 expression in chBMMs and hMDMs compared to C. jejuni WT. Phagocytosis assays showed the acapsular mutant was not impaired in uptake or net intracellular survival after phagocytosis in both chicken and human macrophages; however, the phagocytosis of the MeOPN mutant was significantly decreased in both chicken and human macrophages. In conclusion, differences in the response of macrophages of varying host origin to Campylobacter were detected. The absence of MeOPN modification on the capsule of C. jejuni did not alter the levels of innate cytokine expression in both chicken and human macrophages compared to the 11168H WT, but affected phagocytosis by host macrophages.


Asunto(s)
Infecciones por Campylobacter/veterinaria , Campylobacter jejuni/fisiología , Pollos , Macrófagos/metabolismo , Enfermedades de las Aves de Corral/microbiología , Amidas/metabolismo , Animales , Cápsulas Bacterianas/metabolismo , Médula Ósea , Infecciones por Campylobacter/microbiología , Campylobacter jejuni/genética , Citocinas/metabolismo , Humanos , Monocitos/metabolismo , Mutación , Ácidos Fosfóricos/metabolismo , Ratas
18.
J Biol Chem ; 291(11): 5803-5816, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26786100

RESUMEN

Attaching and effacing Escherichia coli cause diarrhea and typically produce lymphostatin (LifA), an inhibitor of mitogen-activated proliferation of lymphocytes and pro-inflammatory cytokine synthesis. A near-identical factor (Efa1) has been reported to mediate adherence of E. coli to epithelial cells. An amino-terminal region of LifA shares homology with the catalytic domain of the large clostridial toxins, which are retaining glycosyltransferases with a DXD motif involved in binding of a metal ion. Understanding the mode(s) of action of lymphostatin has been constrained by difficulties obtaining a stably transformed plasmid expression clone. We constructed a tightly inducible clone of enteropathogenic E. coli O127:H6 lifA for affinity purification of lymphostatin. The purified protein inhibited mitogen-activated proliferation of bovine T lymphocytes in the femtomolar range. It is a monomer in solution and the molecular envelope was determined using both transmission electron microscopy and small-angle x-ray scattering. Domain architecture was further studied by limited proteolysis. The largest proteolytic fragment containing the putative glycosyltransferase domain was tested in isolation for activity against T cells, and was not sufficient for activity. Tryptophan fluorescence studies indicated thatlymphostatin binds uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc) but not UDP-glucose (UDP-Glc). Substitution of the predicted DXD glycosyltransferase motif with alanine residues abolished UDP-GlcNAc binding and lymphostatin activity, although other biophysical properties were unchanged. The data indicate that lymphostatin has UDP-sugar binding potential that is critical for activity, and is a major leap toward identifying the nature and consequences of modifications of host cell factors.


Asunto(s)
Toxinas Bacterianas/química , Toxinas Bacterianas/inmunología , Enfermedades de los Bovinos/inmunología , Escherichia coli Enteropatógena/inmunología , Infecciones por Escherichia coli/veterinaria , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/inmunología , Linfocitos T/microbiología , Secuencia de Aminoácidos , Animales , Bovinos , Enfermedades de los Bovinos/microbiología , Escherichia coli Enteropatógena/química , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Glicosiltransferasas/química , Glicosiltransferasas/inmunología , Humanos , Activación de Linfocitos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Dispersión del Ángulo Pequeño , Alineación de Secuencia , Linfocitos T/inmunología , Factores de Virulencia/inmunología , Difracción de Rayos X
19.
Infect Immun ; 85(2)2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27920212

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) and enteropathogenic E. coli (EPEC) are enteric bacterial pathogens of worldwide importance. Most EPEC and non-O157 EHEC strains express lymphostatin (also known as LifA), a chromosomally encoded 365-kDa protein. We previously demonstrated that lymphostatin is a putative glycosyltransferase that is important in intestinal colonization of cattle by EHEC serogroup O5, O111, and O26 strains. However, the nature and consequences of the interaction between lymphostatin and immune cells from the bovine host are ill defined. Using purified recombinant protein, we demonstrated that lymphostatin inhibits mitogen-activated proliferation of bovine T cells and, to a lesser extent, proliferation of cytokine-stimulated B cells, but not NK cells. It broadly affected the T cell compartment, inhibiting all cell subsets (CD4, CD8, WC-1, and γδ T cell receptor [γδ-TCR]) and cytokines examined (interleukin 2 [IL-2], IL-4, IL-10, IL-17A, and gamma interferon [IFN-γ]) and rendered T cells refractory to mitogen for a least 18 h after transient exposure. Lymphostatin was also able to inhibit proliferation of T cells stimulated by IL-2 and by antigen presentation using a Theileria-transformed cell line and autologous T cells from Theileria-infected cattle. We conclude that lymphostatin is likely to act early in T cell activation, as stimulation of T cells with concanavalin A, but not phorbol 12-myristate 13-acetate combined with ionomycin, was inhibited. Finally, a homologue of lymphostatin from E. coli O157:H7 (ToxB; L7095) was also found to possess comparable inhibitory activity against T cells, indicating a potentially conserved strategy for interference in adaptive responses by attaching and effacing E. coli.


Asunto(s)
Antígenos Bacterianos/inmunología , Linfocitos B/inmunología , Toxinas Bacterianas/inmunología , Infecciones por Escherichia coli/inmunología , Proteínas de Escherichia coli/inmunología , Escherichia coli/inmunología , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Presentación de Antígeno/inmunología , Linfocitos B/metabolismo , Adhesión Bacteriana , Bovinos , Citocinas/biosíntesis , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Interacciones Huésped-Patógeno , Activación de Linfocitos/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/metabolismo
20.
Microbiology (Reading) ; 163(1): 1-3, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28218576

RESUMEN

of enterohaemorrhagic Escherichia coli O157 pathogenesis using basic symbols. The right-hand illustration shows enterohaemorrhagic E. coli interactions with an epithelial cell.Escherichia coli O157 : H7 is a zoonotic diarrhoeal pathogen of worldwide importance. It belongs to a subset of Shiga toxin-producing E. coli that can form attaching and effacing lesions on intestinal epithelia via the action of a type 3 secretion system that injects bacterial effectors into enterocytes. Infections in humans often arise from contaminated food or direct environmental exposure and can involve life-threatening Shiga toxin-dependent sequelae. In the three decades since E. coli O157 : H7 was first recognized intensive research has helped to unravel the basis of pathogenesis, but few effective options for prevention and treatment of infections exist.


Asunto(s)
Infecciones por Escherichia coli/patología , Escherichia coli O157/patogenicidad , Síndrome Hemolítico-Urémico/microbiología , Mucosa Intestinal/patología , Toxina Shiga/metabolismo , Infecciones por Escherichia coli/microbiología , Escherichia coli O157/genética , Escherichia coli O157/metabolismo , Genoma Bacteriano/genética , Síndrome Hemolítico-Urémico/patología , Humanos , Mucosa Intestinal/microbiología , Sistemas de Secreción Tipo III/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA