Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Microbiol ; 12: 732969, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34646252

RESUMO

Antibiotic-resistant Campylobacter constitutes a serious threat to public health. The clonal expansion of resistant strains and/or the horizontal spread of resistance genes to other strains and species can hinder the clinical effectiveness of antibiotics to treat severe campylobacteriosis. Still, gaps exist in our understanding of the risks of acquisition and spread of antibiotic resistance in Campylobacter. While the in vitro transfer of antimicrobial resistance genes between Campylobacter species via natural transformation has been extensively demonstrated, experimental studies have favored the use of naked DNA to obtain transformants. In this study, we used experimental designs closer to real-world conditions to evaluate the possible transfer of antimicrobial resistance genes between Campylobacter strains of the same or different species (Campylobacter coli or Campylobacter jejuni) and originating from different animal hosts (swine or turkeys). This was evaluated in vitro through co-culture experiments and in vivo with dual-strain inoculation of turkeys, followed by whole genome sequencing of parental and newly emerged strains. In vitro, we observed four independent horizontal gene transfer events leading to the acquisition of resistance to beta-lactams (blaOXA), aminoglycosides [aph(2'')-If and rpsL] and tetracycline [tet(O)]. Observed events involved the displacement of resistance-associated genes by a mutated version, or the acquisition of genomic islands harboring a resistance determinant by homologous recombination; we did not detect the transfer of resistance-carrying plasmids even though they were present in some strains. In vivo, we recovered a newly emerged strain with dual-resistance pattern and identified the replacement of an existing non-functional tet(O) by a functional tet(O) in the recipient strain. Whole genome comparisons allowed characterization of the events involved in the horizontal spread of resistance genes between Campylobacter following in vitro co-culture and in vivo dual inoculation. Our study also highlights the potential for antimicrobial resistance transfer across Campylobacter species originating from turkeys and swine, which may have implications for farms hosting both species in close proximity.

2.
Front Vet Sci ; 8: 613203, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33889603

RESUMO

Consumption of contaminated poultry products is one of the main sources of human campylobacteriosis, of which Campylobacter jejuni subsp. jejuni (C. jejuni) and C. coli are responsible for ~98% of the cases. In turkeys, the ceca are an important anatomical site where Campylobacter asymptomatically colonizes. We previously demonstrated that commercial turkey poults colonized by C. jejuni showed acute changes in cytokine gene expression profiles, and histological intestinal lesions at 2 days post-inoculation (dpi). Cecal tonsils (CT) are an important part of the gastrointestinal-associated lymphoid tissue that surveil material passing in and out of the ceca, and generate immune responses against intestinal pathogens. The CT immune response toward Campylobacter remains unknown. In this study, we generated a kanamycin-resistant C. coli construct (CcK) to facilitate its enumeration from cecal contents after experimental challenge. In vitro analysis of CcK demonstrated no changes in motility when compared to the parent isolate. Poults were inoculated by oral gavage with CcK (5 × 107 colony forming units) or sterile-media (mock-colonized), and euthanized at 1 and 3 dpi. At both time points, CcK was recovered from cecal contents, but not from the mock-colonized group. As a marker of acute inflammation, serum alpha-1 acid glycoprotein was significantly elevated at 3 dpi in CcK inoculated poults compared to mock-infected samples. Significant histological lesions were detected in cecal and CT tissues of CcK colonized poults at 1 and 3 dpi, respectively. RNAseq analysis identified 250 differentially expressed genes (DEG) in CT from CcK colonized poults at 3 dpi, of which 194 were upregulated and 56 were downregulated. From the DEG, 9 significantly enriched biological pathways were identified, including platelet aggregation, response to oxidative stress and negative regulation of oxidative stress-induced intrinsic apoptotic signaling pathway. These data suggest that C. coli induced an acute inflammatory response in the intestinal tract of poults, and that platelet aggregation and oxidative stress in the CT may affect the turkey's ability to resist Campylobacter colonization. These findings will help to develop and test Campylobacter mitigation strategies to promote food safety in commercial turkeys.

3.
Vet Immunol Immunopathol ; 232: 110181, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33401108

RESUMO

Non-typhoidal Salmonella is one of the most common causes of bacterial foodborne disease and consumption of contaminated poultry products, including turkey, is one source of exposure. Minimizing Salmonella colonization of commercial turkeys could decrease the incidence of Salmonella-associated human foodborne illness. Understanding host responses to these bacteria is critical in developing strategies to minimize colonization and reduce food safety risk. In this study, we evaluated bacterial load and blood leukocyte transcriptomic responses of 3-week-old turkeys challenged with the Salmonella enterica serovar Typhimurium (S. Typhimurium) UK1 strain. Turkeys (n = 8/dose) were inoculated by oral gavage with 108 or 1010 colony forming units (CFU) of S. Typhimurium UK1, and fecal shedding and tissue colonization were measured across multiple days post-inoculation (dpi). Fecal shedding was 1-2 log10 higher in the 1010 CFU group than the 108 CFU group, but both doses effectively colonized the crop, spleen, ileum, cecum, colon, bursa of Fabricius and cloaca without causing any detectable clinical signs in either group of birds. Blood leukocytes were isolated from a subset of the birds (n = 3-4/dpi) both pre-inoculation (0 dpi) and 2 dpi with 1010 CFU and their transcriptomic responses assayed by RNA-sequencing (RNA-seq). At 2 dpi, 647 genes had significant differential expression (DE), including large increases in expression of immune genes such as CCAH221, IL4I1, LYZ, IL13RA2, IL22RA2, and ACOD1. IL1ß was predicted as a major regulator of DE in the leukocytes, which was predicted to activate cell migration, phagocytosis and proliferation, and to impact the STAT3 and toll-like receptor pathways. These analyses revealed genes and pathways by which turkey blood leukocytes responded to the pathogen and can provide potential targets for developing intervention strategies or diagnostic assays to mitigate S. Typhimurium colonization in turkeys.


Assuntos
Leucócitos/metabolismo , Doenças das Aves Domésticas/imunologia , Salmonelose Animal/imunologia , Salmonella enterica , Perus , Animais , Leucócitos/imunologia , Masculino , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/microbiologia , Salmonelose Animal/genética , Salmonelose Animal/microbiologia , Transcrição Gênica
4.
Poult Sci ; 99(8): 4028-4033, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32731990

RESUMO

Consumption of contaminated poultry products, including chicken livers, is the main source of human campylobacteriosis and approximately 90% of human cases are caused by Campylobacter jejuni subsp. jejuni (C. jejuni). Recent culinary trends that favor undercooked chicken livers may be responsible for outbreaks. Turkey is an emerging human protein source, and poultry livers are commonly prepared in popular cuisine such as pâté. The mechanism of how Campylobacter disseminates to poultry liver tissue is unknown. We have previously demonstrated that certain strains of C. jejuni persistently colonize turkeys with the highest density in the ceca. Whether C. jejuni disseminates to the liver of turkeys following intestinal colonization is unknown. In this study, 45 D of hatch turkey poults were co-housed for 30 D. Five poults were euthanized to screen for Campylobacter colonization, and were free of detectable Campylobacter. The remaining 40 poults were randomly split into 2 rooms, with 20 poults per room. At 35 D of age, poults were inoculated by oral gavage with 1 × 106 cfu of C. jejuni isolate NCTC 11168 or mock-inoculated with sterile medium. Ten poults from each room were euthanized at 7 and 14 D post-inoculation (dpi), and cecal contents and livers were cultured and/or enriched for Campylobacter. Livers were harvested aseptically. The ceca of C. jejuni-inoculated poults were highly colonized at 7 and 14 dpi with approximately 108 cfu/mL of cecal contents. At 7 and 14 dpi, 3 and 5 of 10 liver samples were positive for C. jejuni culture (8.6 × 103 cfu/g of liver ± 4.43 × 103 and 5.10 × 103 cfu/g of liver ± 1.74 × 103), respectively. At 14 dpi, liver samples were cultured by enrichment, and 6 of 10 were positive for Campylobacter. Some liver samples may be below the limit of detection for direct plate culturing. These data determined that turkey liver is a potential reservoir of C. jejuni following intestinal colonization, and identified a potential food safety consideration when turkey liver is prepared for human or pet food consumption.


Assuntos
Infecções por Campylobacter , Campylobacter jejuni , Fígado , Doenças das Aves Domésticas , Perus , Animais , Infecções por Campylobacter/microbiologia , Infecções por Campylobacter/patologia , Infecções por Campylobacter/veterinária , Campylobacter jejuni/isolamento & purificação , Fígado/microbiologia , Fígado/patologia , Doenças das Aves Domésticas/microbiologia , Doenças das Aves Domésticas/patologia , Distribuição Aleatória
5.
J Anim Sci Biotechnol ; 11: 60, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32537141

RESUMO

BACKGROUND: The initial intestinal microbiota acquired from different sources has profound impacts on animal health and productivity. In modern poultry production practices, the source(s) of the establishing microbes and their overall contribution during development of gastrointestinal tract communities are still unclear. Using fertilized eggs from two independent sources, we assessed the impact of eggshell- and environmental-associated microbial communities on the successional processes and bacterial community structure throughout the intestinal tract of chickens for up to 6 weeks post-hatch. RESULTS: Culturing and sequencing techniques identified a viable, highly diverse population of anaerobic bacteria on the eggshell. The jejunal, ileal, and cecal microbial communities for the egg-only, environment-only, and conventionally raised birds generally displayed similar successional patterns characterized by increasing community richness and evenness over time, with strains of Enterococcus, Romboutsia, and unclassified Lachnospiraceae abundant for all three input groups in both trials. Bacterial community structures differed significantly based on trial and microbiota input with the exception of the egg-exposed and conventional birds in the jejunum at week 1 and the ileum at week 6. Cecal community structures were different based on trial and microbiota input source, and cecal short-chain fatty acid profiles at week 6 highlighted functional differences as well. CONCLUSION: We identified distinct intestinal microbial communities and differing cecal short-chain fatty acid profiles between birds exposed to the microbiota associated with either the eggshell or environment, and those of conventionally hatched birds. Our data suggest the eggshell plays an appreciable role in the development of the chicken intestinal microbiota, especially in the jejunum and ileum where the community structure of the eggshell-only birds was similar to the structure of conventionally hatched birds. Our data identify a complex interplay between the eggshell and environmental microbiota during establishment and succession within the chicken gut. Further studies should explore the ability of eggshell- and environment-derived microbes to shape the dynamics of succession and how these communities can be targeted through interventions to promote gut health and mitigate food-borne pathogen colonization in poultry.

6.
Microbiol Resour Announc ; 9(1)2020 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-31896652

RESUMO

Campylobacter jejuni is the main cause of bacterial foodborne disease in humans, who are exposed mostly by consumption of contaminated poultry products. C. jejuni strain NADC 20827 was isolated from the feces of turkeys naturally colonized with Campylobacter spp. We present the complete annotated genome and plasmid sequences of strain NADC 20827.

7.
Microorganisms ; 7(10)2019 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-31547217

RESUMO

The loss of antibiotics as a tool to improve feed efficiency in poultry production has increased the urgency to understand how the microbiota interacts with animals to impact productivity and health. Modulating and harnessing microbiota-host interactions is a promising way to promote poultry health and production efficiencies without antibiotics. In poultry, the microbiome is influenced by many host and external factors including host species, age, gut compartment, diet, and environmental exposure to microbes. Because so many factors contribute to the microbiota composition, specific knowledge is needed to predict how the microbiome will respond to interventions. The effects of antibiotics on microbiomes have been well documented, with different classes of antibiotics having distinctive, specific outcomes on bacterial functions and membership. Non-antibiotic interventions, such as probiotics and prebiotics, target specific bacterial taxa or function to enhance beneficial properties of microbes in the gut. Beneficial bacteria provide a benefit by displacing pathogens and/or producing metabolites (e.g., short chain fatty acids or tryptophan metabolites) that promote poultry health by improving mucosal barrier function or immune function. Microbiota modulation has been used as a tool to reduce pathogen carriage, improve growth, and modulate the immune system. An increased understanding of how the microbiota interacts with animal hosts will improve microbiome intervention strategies to mitigate production losses without the need for antibiotics.

8.
Sci Rep ; 9(1): 8212, 2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31160613

RESUMO

Beginning in 2017, the subtherapeutic use of most antibiotic compounds for growth promotion in food producing animals in the US was prohibited, highlighting the need to discover alternative growth promotants. Identifying the mechanism of action of growth promoting antibiotics may aid in the discovery of antibiotic alternatives. We describe the effects of feeding a subtherapeutic (50 g/ton of feed) and therapeutic (200 g/ton) concentration of bacitracin methylene disalicylate (BMD) to commercial turkeys for 14 weeks, and its effect on turkey intestinal microbial communities and cecal metabolomes. Both BMD concentrations had an immediate and lasting impact on the microbiota structure, and reduced bacterial richness through the end of the study (12 weeks). Metabolomic analysis identified 712 biochemicals, and 69% of metabolites were differentially present in BMD treated turkeys for at least one time point (q < 0.1). Amino acids, carbohydrates, nucleotides, peptides, and lipids were decreased in the turkey ceca early after BMD administration. Long-term metabolome alterations continued even after withdrawal of BMD. The microbial composition, determined by 16S rRNA gene sequencing, was predictive of the metabolome, indicating a connection between the microbiome and metabolome. In-feed BMD may cause bacterial metabolic shifts, leading to beneficial traits that can be targeted to improve animal health and production.


Assuntos
Ração Animal/análise , Bacitracina/farmacologia , Metaboloma/efeitos dos fármacos , Microbiota/efeitos dos fármacos , Salicilatos/farmacologia , Perus/metabolismo , Perus/microbiologia , Aminoácidos/análise , Animais , Bactérias/efeitos dos fármacos , Biodiversidade , Ceco/microbiologia , Masculino , Fatores de Tempo
9.
Vet Immunol Immunopathol ; 210: 6-14, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30947981

RESUMO

Consumption of contaminated poultry products is one of the main sources of human campylobacteriosis, of which Campylobacter jejuni subsp. jejuni (C. jejuni) is responsible for approximately 90% of the cases. At slaughter, the ceca of commercial chickens and turkeys are the main anatomical site where C. jejuni asymptomatically colonizes. We have previously colonized commercial turkey poults with different isolates of C. jejuni and evaluated different media to best enumerate Campylobacter from intestinal samples, but the host-response is unknown in turkeys. Enumeration of Campylobacter (colony forming units (cfu)/gram of intestinal contents) can be challenging, and can be confounded if animals are colonized with multiple species of Campylobacter. In order to precisely enumerate the C. jejuni isolate used to experimentally colonize turkeys, constructs of C. jejuni (NCTC 11,168) were tagged with different antibiotic resistance markers at the CmeF locus (chloramphenicol (CjCm) or kanamycin (CjK)). We sought to examine the kinetics of intestinal colonization using the antibiotic resistant constructs, and characterize the immune response in cecal tissue of turkeys. In vitro analysis of the tagged antibiotic-resistant constructs demonstrated no changes in motility, morphology, or adherence and invasion of INT-407 cells compared to the parent isolate NCTC 11,168. Two animal experiments were completed to evaluate intestinal colonization by the constructs. In experiment 1, three-week old poults were colonized after oral gavage for 14 days, and CjCm and CjK cfu were recovered from cecal, but not ileal contents. In experiment 2, nine-week old poults were orally inoculated with CjCm, and the abundance of CjCm cfu/g of cecal contents significantly decreased beyond 14 days after inoculation. Significant lesions were detected in CjCm colonized poults at day 2 post-colonization. Using immunohistochemistry, Campylobacter antigen was detected in between cecal villi by day 7 of CjCm colonized poults. Quantitative RT-PCR of CjCm-colonized cecal tissue demonstrated significant down-regulation of IL-1ß, IL-10 and IL-13 mRNA, and significant up-regulation of IL-6, IL-8, IL-17 A, IL-22 and IFNγ mRNA on day 2, and for some on day 7 post-colonization. All differentially expressed genes were similar to mock-infected poults by day 14. These data suggest that C. jejuni induced a brief inflammatory response in the cecum of poults that quickly resolved. Results from this study provide valuable insight into host-response and persistent colonization of the turkey cecum. These findings will help to develop and test strategies to promote food safety in commercial turkeys.


Assuntos
Infecções por Campylobacter/veterinária , Campylobacter jejuni/genética , Resistência Microbiana a Medicamentos/genética , Mucosa Intestinal/imunologia , Intestinos/microbiologia , Animais , Infecções por Campylobacter/imunologia , Ceco/imunologia , Ceco/microbiologia , Contagem de Colônia Microbiana , Citocinas/genética , Microbiologia de Alimentos , Marcadores Genéticos , Interações entre Hospedeiro e Microrganismos/imunologia , Íleo/imunologia , Íleo/microbiologia , Inflamação , Intestinos/imunologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/microbiologia , Perus , Vacinação/veterinária
10.
Vaccine ; 37(10): 1255-1259, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30718082

RESUMO

Salmonella vaccine strategies for food-producing animals have typically focused on a specific serovar that either causes production losses due to morbidity/mortality or is an important food safety pathogen for a particular food commodity. The Salmonella enterica serovar Typhimurium BBS 866 vaccine strain was designed to reduce serovar specificity to provide cross-protection against diverse Salmonella serovars, thereby broadening its applicability for multiple animal and poultry species. We reported cross-protection of the BBS 866 vaccine in swine [Vaccine 34:1241-6]. In the current study, we extend the efficacy of the Salmonella vaccine to a poultry commodity by revealing significant reduction of multidrug-resistant Salmonella enterica serovar Heidelberg colonization of the cecum and systemic dissemination to the spleen in BBS 866-vaccinated turkeys. Transcriptional analysis of whole blood from BBS 866-vaccinated turkeys revealed down-regulation of metabolic and immune genes (KCNAB1, ACOD1, GPR17, ADOR2AB, and IL-17RD), suggesting limited leukocyte activation without an overt peripheral inflammatory response to vaccination.


Assuntos
Ceco/microbiologia , Doenças das Aves Domésticas/prevenção & controle , Salmonelose Animal/prevenção & controle , Vacinas contra Salmonella/imunologia , Salmonella enterica/crescimento & desenvolvimento , Baço/microbiologia , Animais , Ceco/imunologia , Proteção Cruzada , Farmacorresistência Bacteriana Múltipla , Expressão Gênica , Masculino , Vacinas contra Salmonella/administração & dosagem , Sorogrupo , Baço/imunologia , Perus/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
11.
Front Microbiol ; 9: 1991, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30197636

RESUMO

Segmented filamentous bacteria (SFB) are a group of host-adapted, commensal organisms that attach to the ileal epithelium of vertebrate and invertebrate hosts. A genetic relative of the genus Clostridium, these morphologically unique bacteria display a replication and differentiation lifecycle initiated by epithelial tissue binding and filamentation. SFB intimately bind to the surface of absorptive intestinal epithelium without inducing an inflammatory response. Rather, their presence impacts the generation of innate and differentiation of acquired immunity, which impact the clearance of extracellular bacterial or fungal pathogens in the gastrointestinal and respiratory tracts. SFB have recently garnered attention due to their role in promoting adaptive and innate immunity in mice and rats through the differentiation and maturation of Th17 cells in the intestinal tract and production of immunoglobulin A (IgA). SFB are the first commensal bacteria identified that impact the maturation and development of Th17 cells in mice. Recently, microbiome studies have revealed the presence of Candidatus Arthromitus (occasionally designated as Candidatus Savagella), a proposed candidate species of SFB, in higher proportions in higher-performing flocks as compared to matched lower-performing flocks, suggesting that SFB may serve to establish a healthy gut and protect commercial turkeys from pathogens resulting in morbidity and decreased performance. In this review we seek to describe the life cycle, host specificity, and genetic capabilities of SFB, such as bacterial metabolism, and how these factors influence the host immunity and microbiome. Although the role of SFB to induce antigen-specific Th17 cells in poultry is unknown, they may play an important role in modulating the immune response in the intestinal tract to promote resistance against some infectious diseases and promote food-safety. This review demonstrates the importance of studying and further characterizing commensal, host-specific bacteria in food-producing animals and their importance to animal health.

12.
Front Microbiol ; 9: 3215, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30687245

RESUMO

Campylobacter jejuni is a leading cause of bacterial foodborne illness in humans worldwide. However, C. jejuni naturally colonizes poultry without causing pathology where it resides deep within mucus of the cecal crypts. Mucus may modulate the pathogenicity of C. jejuni in a species-specific manner, where it is pathogenic in humans and asymptomatic in poultry. Little is known about how intestinal mucus from different host species affects C. jejuni gene expression. In this study we characterized the growth and transcriptome of C. jejuni NCTC11168 cultured in defined media supplemented with or without mucus isolated from avian (chicken or turkey) or mammalian (cow, pig, or sheep) sources. C. jejuni showed substantially improved growth over defined media, with mucus from all species, showing that intestinal mucus was an energy source for C. jejuni. Seventy-three genes were differentially expressed when C. jejuni was cultured in avian vs. mammalian mucus. Genes associated with iron acquisition and resistance to oxidative stress were significantly increased in avian mucus. Many of the differentially expressed genes were flanked by differentially expressed antisense RNA asRNA, suggesting a role in gene regulation. This study highlights the interactions between C. jejuni and host mucus and the impact on gene expression, growth and invasion of host cells, suggesting important responses to environmental cues that facilitate intestinal colonization. IMPORTANCE  Campylobacter jejuni infection of humans is an important health problem world-wide and is the leading bacterial cause of foodborne illnesses in U.S. The main route for exposure for humans is consumption of poultry meat contaminated during processing. C. jejuni is frequently found in poultry, residing within the mucus of the intestinal tract without causing disease. It is not clear why C. jejuni causes disease in some animals and humans, while leaving birds without symptoms. To understand its activity in birds, we characterized C. jejuni responses to poultry mucus to identify genes turned on in the intestinal tract of birds. We identified genes important for colonization and persistence within the poultry gut, turned on when C. jejuni was exposed to poultry mucus. Our findings are an important step in understanding how C. jejuni responds and interacts in the poultry gut, and may identify ways to reduce C. jejuni in birds.

13.
Vet Immunol Immunopathol ; 191: 74-79, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28895870

RESUMO

Between December 2014 and June 2015, North America experienced the largest recorded foreign animal disease outbreak with over 47 million poultry dead or euthanized from viral exposure to a clade 2.3.4.4 H5 highly pathogenic avian influenza (HPAI) epizootic. Soon after the epizootic began, the U.S. Department of Agriculture (USDA) began testing the efficacy of different vaccines as a possible future control strategy. The aim of these studies were to evaluate the efficacy three H5 vaccines to aid in control of HPAI in commercial turkeys. Three different vaccine technologies were evaluated for efficacy: 1) inactivated reverse genetic laboratory-generated virus encoding a clade 2.3.4.4 H5 hemagglutinin (HA) gene (rgH5), 2) recombinant turkey herpesvirus encoding a clade 2.2. H5 HA (rHVT-AI), and 3) recombinant replication-deficient alphavirus RNA particle vaccine encoding a clade 2.3.4.4 H5 HA (RP-H5). All vaccines tested significantly (P<0.01) increased survival rates between vaccinated and sham vaccinated groups of poults challenged with A/turkey/Minnesota/12582/2015 clade 2.3.4.4 H5N2 HPAI. The rgH5 vaccine had detectable serum hemagglutination inhibition (HI) antibody against the challenge virus, and significantly reduced the frequency and level of viral shedding from oropharyngeal and cloacal swabs at days 2 and 4 post-challenge. Vaccination with only rHVT-AI or RP-H5 was not 100% protective, and failed to significantly reduce viral shedding post-challenge. A combined prime and boost strategy with the rHVT-AI and RP-H5, or rHVT-AI and rgH5, was 100% protective against lethal H5N2 HPAI challenge. Results of these studies led to USDA conditional approval of commercially available recombinant vaccines for use in turkeys as a control measure for clade 2.3.4.4 H5 HPAI epizootics.


Assuntos
Vírus da Influenza A Subtipo H5N2/imunologia , Vacinas contra Influenza/uso terapêutico , Influenza Aviária/prevenção & controle , Perus/virologia , Animais , Feminino , Vírus da Influenza A Subtipo H5N2/patogenicidade , Vacinas contra Influenza/imunologia , Influenza Aviária/imunologia , Influenza Aviária/virologia , Masculino , Perus/imunologia , Vacinas de Produtos Inativados/imunologia , Vacinas de Produtos Inativados/uso terapêutico , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico , Eliminação de Partículas Virais/imunologia
14.
Vaccine ; 30(20): 3097-105, 2012 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-22394994

RESUMO

Methods to determine vaccine efficacy of low pathogenic avian influenza (LPAI) isolates are limited in poultry because experimental infections with LPAI virus in specific pathogen free chickens rarely causes clinical disease. The most commonly used method to compare LPAI vaccine efficacy is to quantify viral shedding after challenge, but it is time and labor intensive. Therefore, we sought alternative methods to demonstrate vaccine efficacy, and examined whether vaccination of chickens affected the production of acute phase mediators in serum (e.g., alpha-1 acid glycoprotein, ovotransferrin and prostaglandin E(2)) following challenge with homologous (A/Chicken/Hidalgo/232/94 H5N2) or heterologous (A/Chicken/167280-4/02 H5N3) LPAI isolates. Vaccination significantly reduced oropharyngeal viral shedding, and serum concentration of acute phase mediators, regardless of whether homologous or heterologous challenge viruses were used. Examining the expression of acute phase mediators post-challenge may serve as additional markers to determine LPAI vaccine efficacy in chickens.


Assuntos
Proteínas de Fase Aguda/metabolismo , Biomarcadores/sangue , Vírus da Influenza A Subtipo H5N2/imunologia , Vacinas contra Influenza/imunologia , Influenza Aviária/prevenção & controle , Influenza Aviária/virologia , Animais , Galinhas , Vírus da Influenza A Subtipo H5N2/patogenicidade , Vacinas contra Influenza/administração & dosagem , Influenza Aviária/imunologia , Orofaringe/virologia , Eliminação de Partículas Virais
15.
Curr Top Microbiol Immunol ; 333: 227-41, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19768409

RESUMO

The neuraminidase protein of influenza viruses is a surface glycoprotein that shows enzymatic activity to remove sialic acid, the viral receptor, from both viral and host proteins. The removal of sialic acid from viral proteins plays a key role in the release of the virus from the cell by preventing the aggregation of the virus by the hemagglutinin protein binding to other viral proteins. Antibodies to the neuraminidase protein can be protective alone in animal challenge studies, but the neuraminidase antibodies appear to provide protection in a different manner than antibodies to the hemagglutinin protein. Neutralizing antibodies to the hemagglutinin protein can directly block virus entry, but protective antibodies to the neuraminidase protein are thought to primarily aggregate virus on the cell surface, effectively reducing the amount of virus released from infected cells. The neuraminidase protein can be divided into nine distinct antigenic subtypes, where there is little cross-protection of antibodies between subtypes. All nine subtypes of neuraminidase protein are commonly found in avian influenza viruses, but only selected subtypes are routinely found in mammalian influenza viruses; for example, only the N1 and N2 subtypes are commonly found in both humans and swine. Even within a subtype, the neuraminidase protein can have a high level of antigenic drift, and vaccination has to specifically be targeted to the circulating strain to give optimal protection. The levels of neuraminidase antibody also appear to be critical for protection, and there is concern that human influenza vaccines do not include enough neuraminidase protein to induce a strong protective antibody response. The neuraminidase protein has also become an important target for antiviral drugs that target sialic acid binding which blocks neuraminidase enzyme activity. Two different antiviral drugs are available and are widely used for the treatment of seasonal influenza in humans, but antiviral resistance appears to be a growing concern for this class of antivirals.


Assuntos
Vacinas contra Influenza/imunologia , Neuraminidase/imunologia , Animais , Antivirais/farmacologia , Farmacorresistência Viral , Inibidores Enzimáticos/farmacologia , Humanos , Neuraminidase/antagonistas & inibidores , Neuraminidase/química , Neuraminidase/fisiologia
16.
Vaccine ; 25(19): 3763-72, 2007 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-17350145

RESUMO

Protection of chickens against avian influenza (AI) is mostly attributed to production of antibodies against the viral glycoprotein hemagglutinin, whereas less is known about the protective role of antibodies to the other surface glycoprotein neuraminidase (NA). Therefore, vaccines encoding NA antigen (e.g., DNA and alphavirus-based virus like replicon particles (VRP)) or baculovirus-expressed recombinant NA (rN2) were tested for their ability to protect against highly pathogenic AI (HPAI) in chickens. Vaccination with A/Pheasant/Maryland/4457/93 (Ph/MD) rN2 protein produced significantly higher levels of NA-inhibition (NI) activity and 88% protection from HPAI H5N2 challenge than vaccination with Ph/MD N2 DNA (25% protection). Vaccination with Ph/MD N2 VRP a minimum of two times also produced high levels of NI activity and protection against HPAI challenge (63% protection). Vaccination with VRP encoding an N2 gene that was genetically distant from the challenge virus N2 failed to protect chickens. Vaccines producing higher levels of NI activity conferred partial protection, but failed to affect viral shedding. Consideration of the homology between vaccine and challenge virus isolate NA genes may provide improved immunity if high levels of NI activity are obtained.


Assuntos
Anticorpos Antivirais , Vírus da Influenza A Subtipo H5N2/patogenicidade , Vacinas contra Influenza/imunologia , Influenza Aviária/prevenção & controle , Neuraminidase/imunologia , Vacinas de DNA/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Galinhas , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/genética , Influenza Aviária/imunologia , Influenza Aviária/mortalidade , Influenza Aviária/virologia , Neuraminidase/administração & dosagem , Neuraminidase/genética , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/mortalidade , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vacinação/veterinária , Vacinas de DNA/administração & dosagem , Eliminação de Partículas Virais
17.
Nutr Cancer ; 50(2): 221-31, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15623470

RESUMO

Low selenium (Se) status increases angiogenesis by inducing the production of vascular endothelial growth factor (VEGF); however, the mechanism responsible for VEGF up-regulation has yet to be characterized. Se's ability to control cellular oxidative state through its incorporation into selenoproteins such as thioredoxin reductase (TrxR) may explain previous studies that connect Se status to tumor angiogenesis. Therefore, the focus of this study was to determine if altered VEGF expression and angiogenesis due to decreased Se levels are influenced by reduced TrxR activity. We found that chemical inhibition of TrxR in Se-sufficient endothelial cells (ECs) was associated with increases in VEGF and VEGF receptor expression, cell migration, proliferation, and angiogenesis to levels similar to those seen in Se-deficient ECs. Specific inhibition of glutathione peroxidase did not affect pro-angiogenic responses, indicating a unique role of the TrxR system during low Se status. These data correlate changes in TrxR activity with changes in VEGF expression and angiogenic development in ECs, which is significant because minimal mechanistic data exist that explain the role of Se in cancer prevention. Understanding the importance of the tumor microenvironment in contributing to angiogenic regulation has the potential to significantly impact breast cancer chemoprevention strategies by focusing on maintaining proper EC function within the mammary gland.


Assuntos
Células Endoteliais/metabolismo , Neovascularização Fisiológica , Tiorredoxina Dissulfeto Redutase/fisiologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Bovinos , Movimento Celular , Proliferação de Células , Células Cultivadas , Glutationa Peroxidase/fisiologia , RNA Mensageiro/análise , Selênio/fisiologia , Tiorredoxina Dissulfeto Redutase/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA