Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Food Microbiol ; 105: 104007, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35473970

RESUMO

Listeria (L.) monocytogenes is a significant pathogen found in ready-to-eat meat and dairy products. Soft cheeses, such as Queso Fresco cheese (QFC), are particularly sensitive to Listeria contamination, and occasionally serve as a source of food-borne illness outbreaks. In the present study, clinical and cheese isolates of L. monocytogenes were assayed for phenotypic characteristics following sub-lethal high voltage atmospheric cold plasma (HVACP) treatment. Reductions in biofilm formation, swimming motility, and growth dynamics were observed following HVACP treatment. Microbial enumeration of 1-, 10-, and 100-g fresh QFC following 0, 1, 2, or 3 min of HVACP demonstrated significant reductions in L. monocytogenes after 1 min (P-value <0.05), with increasing efficacy with prolonged exposure. A mass-dependent effect was observed between treatments of 1-, 10-, and 100-g QFC in regard to treatment efficacy. This result indicates that greater L. monocytogenes reduction on a larger QFC mass requires greater exposure of the L. monocytogenes to the reactive gas species. Optical absorption spectroscopy confirmed a reduction in reactive gas species for each log increase in QFC mass, however, an equivalent volume of inert foam resulted in increased reactive gas generation compared to QFC. In conclusion, we demonstrate both the application and limitations of HVACP treatments of QFC in the currently defined experimental parameters.


Assuntos
Queijo , Listeria monocytogenes , Listeria , Gases em Plasma , Microbiologia de Alimentos , Gases em Plasma/farmacologia
2.
Food Microbiol ; 95: 103669, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33397632

RESUMO

Fungal contamination is a concern for the food industry. Fungal spores resist food sterilization treatments and produce mycotoxins that are toxic for animals and humans. Technologies that deactivate spores and toxins without impacting food quality are desirable. This study demonstrates the efficiency of a high voltage atmospheric cold plasma (HVACP) technology using air to generate reactive oxygen (ROS) and nitrogen (RNS) species for the degradation of Aspergillus flavus cultures and the deoxynivalenol (DON) mycotoxin. Optical emission and absorption spectroscopy demonstrate ionization of hydroxyl groups, atomic oxygen and nitrogen, and confirm production of ROS and RNS, e.g. O3, NO2, NO3, N2O4, and N2O5. Fungal cultures show a depletion in pigmentation and an ~50% spore inactivation after 1-min treatments. Treated spores show surface ablation and membrane degradation by scanning electron microscopy. Twenty-minute direct HVACP treatments of 100 µg of DON in one mL aqueous suspensions resulted in a greater than 99% reduction in DON structure and rescued over 80% of Caco-2 cell viability; however, the same treatment on 100 µg of powdered DON toxin only showed a 33% reduction in DON and only rescued 15% of cell viability. In summary, HVACP air treatment can inactivate both fungal spores and toxins in minutes.


Assuntos
Aspergillus flavus/efeitos dos fármacos , Gases em Plasma/farmacologia , Esporos Fúngicos/crescimento & desenvolvimento , Tricotecenos/metabolismo , Aspergilose/microbiologia , Aspergillus flavus/crescimento & desenvolvimento , Aspergillus flavus/metabolismo , Células CACO-2 , Humanos , Gases em Plasma/química , Espécies Reativas de Oxigênio/metabolismo , Esporos Fúngicos/efeitos dos fármacos , Esporos Fúngicos/metabolismo
3.
Foodborne Pathog Dis ; 15(3): 119-131, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29638166

RESUMO

Cattle are a major reservoir for Shiga toxin-producing Escherichia coli (STEC) and harbor these bacteria in the intestinal tract. The prevalence, concentration, and STEC serogroup isolated in cattle varies between individuals. Hide removal at slaughter serves as a major point of carcass contamination and ultimately beef products. Certain STEC serogroups, such as O26, O45, O103, O111, O121, O145, and O157, containing the intestinal adherence factor intimin, pose a large economic burden to food producers because of testing and recalls. Human infection with STEC can cause illnesses ranging from diarrhea to hemorrhagic colitis and hemolytic uremic syndrome, and is commonly acquired through ingestion of contaminated foods, often beef products. Previously, most studies focused on O157 STEC, but there is growing recognition of the importance of non-O157 STEC serogroups. This review summarizes detection methods, prevalence, and methods for prediction of pathogenicity of non-O157 STEC from cattle hides and carcasses. A synthesis of procedures is outlined for general non-O157 STEC and targeted detection of specific STEC serogroups. Standardization of sample collection and processing procedures would allow for more robust comparisons among studies. Presence of non-O157 STEC isolated from cattle hides and carcasses and specific factors, such as point of sample collection and season, are summarized. Also, factors that might influence STEC survival on these surfaces, such as the microbial population on hides and microbial adherence genes, are raised as topics for future investigation. Finally, this review gives an overview on studies that have used genetic and cell-based methods to identify specific phenotypes of non-O157 STEC strains isolated from cattle to assess their risk to human health.


Assuntos
Reservatórios de Doenças/microbiologia , Infecções por Escherichia coli/epidemiologia , Síndrome Hemolítico-Urêmica/epidemiologia , Carne Vermelha/microbiologia , Escherichia coli Shiga Toxigênica/imunologia , Animais , Bovinos , Diarreia/epidemiologia , Diarreia/microbiologia , Diarreia/prevenção & controle , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/prevenção & controle , Síndrome Hemolítico-Urêmica/microbiologia , Síndrome Hemolítico-Urêmica/prevenção & controle , Humanos , Fenótipo , Prevalência , Estações do Ano , Sorogrupo , Escherichia coli Shiga Toxigênica/isolamento & purificação , Escherichia coli Shiga Toxigênica/patogenicidade , Virulência
4.
Appl Environ Microbiol ; 81(3): 1177-87, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25480753

RESUMO

Chicken products are suspected as a source of extraintestinal pathogenic Escherichia coli (ExPEC), which causes diseases in humans. The zoonotic risk to humans from chicken-source E. coli is not fully elucidated. To clarify the zoonotic risk posed by ExPEC in chicken products and to fill existing knowledge gaps regarding ExPEC zoonosis, we evaluated the prevalence of ExPEC on shell eggs and compared virulence-associated phenotypes between ExPEC and non-ExPEC isolates from both chicken meat and eggs. The prevalence of ExPEC among egg-source isolates was low, i.e., 5/108 (4.7%). Based on combined genotypic and phenotypic screening results, multiple human and avian pathotypes were represented among the chicken-source ExPEC isolates, including avian-pathogenic E. coli (APEC), uropathogenic E. coli (UPEC), neonatal meningitis E. coli (NMEC), and sepsis-associated E. coli (SEPEC), as well as an undefined ExPEC group, which included isolates with fewer virulence factors than the APEC, UPEC, and NMEC isolates. These findings document a substantial prevalence of human-pathogenic ExPEC-associated genes and phenotypes among E. coli isolates from retail chicken products and identify key virulence traits that could be used for screening.


Assuntos
Ovos/microbiologia , Infecções por Escherichia coli/transmissão , Escherichia coli/isolamento & purificação , Produtos da Carne/microbiologia , Zoonoses/transmissão , Animais , Galinhas , Escherichia coli/classificação , Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Genótipo , Humanos , Fenótipo , Reação em Cadeia da Polimerase , Prevalência , Medição de Risco , Fatores de Virulência/genética , Zoonoses/microbiologia
5.
Front Microbiol ; 15: 1414401, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38903782

RESUMO

The animal gut acts as a potent reservoir for spreading and maintaining conjugative plasmids that confer antimicrobial resistance (AMR), fitness, and virulence attributes. Interventions that inhibit the continued emergence and expansion of AMR and virulent strains in agricultural and clinical environments are greatly desired. This study aims to determine the presence and efficacy of short-chain fatty acids (SCFA) inhibitory effects on the conjugal transfer of AMR plasmids. In vitro broth conjugations were conducted between donor Escherichia coli strains carrying AMP plasmids and the plasmid-less Escherichia coli HS-4 recipient strain. Conjugations were supplemented with ddH2O or SCFAs at 1, 0.1, 0.01, or 0.001 molar final concentration. The addition of SCFAs completely inhibited plasmid transfer at 1 and 0.1 molar and significantly (p < 0.05) reduced transfer at 0.01 molar, regardless of SCFA tested. In explant models for the chicken ceca, either ddH2O or a final concentration of 0.025 M SCFAs were supplemented to the explants infected with donor and recipient E. coli. In every SCFA tested, significant decreases in transconjugant populations compared to ddH2O-treated control samples were observed with minimal effects on donor and recipient populations. Finally, significant reductions in transconjugants for plasmids of each incompatibility type (IncP1ε, IncFIß, and IncI1) tested were detected. This study demonstrates for the first time the broad inhibition ability of SCFAs on bacterial plasmid transfer and eliminates AMR with minimal effect on bacteria. Implementing interventions that increase the concentrations of SCFAs in the gut may be a viable method to reduce the risk, incidence, and rate of AMR emergence in agricultural and human environments.

6.
Infect Immun ; 81(3): 838-49, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23275093

RESUMO

Avian pathogenic Escherichia coli (APEC) causes respiratory and systemic disease in poultry. Sequencing of a multilocus sequence type 95 (ST95) serogroup O1 strain previously indicated that APEC resembles E. coli causing extraintestinal human diseases. We sequenced the genomes of two strains of another dominant APEC lineage (ST23 serogroup O78 strains χ7122 and IMT2125) and compared them to each other and to the reannotated APEC O1 sequence. For comparison, we also sequenced a human enterotoxigenic E. coli (ETEC) strain of the same ST23 serogroup O78 lineage. Phylogenetic analysis indicated that the APEC O78 strains were more closely related to human ST23 ETEC than to APEC O1, indicating that separation of pathotypes on the basis of their extraintestinal or diarrheagenic nature is not supported by their phylogeny. The accessory genome of APEC ST23 strains exhibited limited conservation of APEC O1 genomic islands and a distinct repertoire of virulence-associated loci. In light of this diversity, we surveyed the phenotype of 2,185 signature-tagged transposon mutants of χ7122 following intra-air sac inoculation of turkeys. This procedure identified novel APEC ST23 genes that play strain- and tissue-specific roles during infection. For example, genes mediating group 4 capsule synthesis were required for the virulence of χ7122 and were conserved in IMT2125 but absent from APEC O1. Our data reveal the genetic diversity of E. coli strains adapted to cause the same avian disease and indicate that the core genome of the ST23 lineage serves as a chassis for the evolution of E. coli strains adapted to cause avian or human disease via acquisition of distinct virulence genes.


Assuntos
Evolução Biológica , Escherichia coli/classificação , Escherichia coli/genética , Genoma Bacteriano/genética , Doenças das Aves Domésticas/microbiologia , Perus , Animais , DNA Bacteriano/genética , Escherichia coli/patogenicidade , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/metabolismo , Regulação Bacteriana da Expressão Gênica , Lactoferrina/deficiência , Transtornos Leucocíticos , Anotação de Sequência Molecular , Dados de Sequência Molecular , Mutação , Filogenia , Virulência
7.
Dis Aquat Organ ; 106(2): 129-38, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-24113246

RESUMO

Flavobacterium columnare, a fastidious Gram-negative pathogen and the causative agent of columnaris disease, is one of the most harmful pathogens in the freshwater fish-farming industry. Nevertheless the virulence mechanisms of F. columnare are not well understood. Bacterial iron uptake from the host during infection is an important mechanism of virulence. Here we identified and analyzed part of the iron uptake machinery of F. columnare. Under iron-limited conditions during in vitro growth, synthesis of an outer membrane protein of ~86 kDa was upregulated. This protein was identified as a TonB-dependent ferrichrome-iron receptor precursor (FhuA). Synthesis of siderophores in F. columnare was corroborated by chrome azurol S assays. A putative ferric uptake regulator (Fur) protein was also identified in the F. columnare genome. Structural analysis of the F. columnare Fur protein revealed that it was similar to Fur proteins involved in iron uptake regulation of other bacteria. Furthermore, Salmonella enterica serovar Typhimurium (S. Typhimurium) Δfur mutants were partially complemented by the F. columnare fur gene. We conclude that a siderophore-mediated iron uptake system exists in F. columnare, and fur from F. columnare could partially complement S. Typhimurium Δfur mutant.


Assuntos
Flavobacterium/metabolismo , Ferro/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Transporte Biológico/fisiologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Sideróforos
8.
Foodborne Pathog Dis ; 10(11): 916-32, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23962019

RESUMO

Extraintestinal pathogenic Escherichia coli (ExPEC) constitutes ongoing health concerns for women, newborns, elderly, and immunocompromised individuals due to increased numbers of urinary tract infections (UTIs), newborn meningitis, abdominal sepsis, and septicemia. E. coli remains the leading cause of UTIs, with recent investigations reporting the emergence of E. coli as the predominant cause of nosocomial and neonatal sepsis infections. This shift from the traditional Gram-positive bacterial causes of nosocomial and neonatal sepsis infections could be attributed to the use of intrapartum chemoprophylaxis against Gram-positive bacteria and the appearance of antibiotic (ATB) resistance in E. coli. While ExPEC strains cause significant healthcare concerns, these bacteria also infect chickens and cause the poultry industry economic losses due to costs of containment, mortality, and disposal of carcasses. To circumvent ExPEC-related costs, ATBs are commonly used in the poultry industry to prevent/treat microbial infections and promote growth and performance. In an unfortunate linkage, chicken products are suspected to be a source of foodborne ExPEC infections and ATB resistance in humans. Therefore, the emergence of multidrug resistance (MDR) (resistance to three or more classes of antimicrobial agents) among avian E. coli has created major economic and health concerns, affecting both human healthcare and poultry industries. Increased numbers of immunocompromised individuals, including the elderly, coupled with MDR among ExPEC strains, will continue to challenge the treatment of ExPEC infections and likely lead to increased treatment costs. With ongoing complications due to emerging ATB resistance, novel treatment strategies are necessary to control ExPEC infections. Recognizing and treating the zoonotic risk posed by ExPEC would greatly enhance food safety and positively impact human health.


Assuntos
Farmacorresistência Bacteriana Múltipla , Infecções por Escherichia coli/diagnóstico , Escherichia coli/isolamento & purificação , Zoonoses/microbiologia , Animais , Celulite (Flegmão)/microbiologia , Celulite (Flegmão)/veterinária , Galinhas/microbiologia , Infecção Hospitalar , Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Feminino , Humanos , Meningite devida a Escherichia coli , Infecções Respiratórias/microbiologia , Infecções Respiratórias/veterinária , Salpingite/microbiologia , Salpingite/veterinária , Sepse/diagnóstico , Sepse/microbiologia , Infecções Urinárias/microbiologia , Fatores de Virulência
9.
Front Microbiol ; 14: 1231837, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37583515

RESUMO

Introduction: Gut microbes like segmented filamentous bacteria (SFB) play a key role in gut maturation during early life, as demonstrated in humans and mice. Our previous study demonstrated oral inoculation of ileum-spores containing SFB to chickens after hatch increases early SFB gut colonization, which increases immune maturation and resistance to bacteria, like Salmonella, as tested in vitro; however, more studies are needed for treatment optimization and in vivo testing. The objectives of this study were to (1) test a treatment that includes both spores and filamentous SFB, (2) validate antimicrobial ability of the treatment in layer hens in vivo, and (3) elucidate its molecular mechanism. Methods: One-day-old specific pathogen-free layers (n = 12 per group) were orally treated with either PBS (CON) or SFB-based treatment (SFB). At 4 days post-inoculation (DPI), both CON and SFB groups were orally challenged with Salmonella Typhimurium. Total Enterobacteriaceae and Salmonella were examined by plating and enumeration in feces at 7,10 and 14 dpi; and in the ileum, cecum, and spleen at 16 dpi in euthanized birds. The presence and levels of SFB were determined from ilea scrapings via microscopy and qPCR, respectively. Relative gene expression of host-derived antimicrobial peptides and cytokines in the distal ileum was determined by RT-qPCR. Results: At 10 and 14 dpi, a significant decrease in total Enterobacteriaceae was observed in the feces of the SFB group. At necropsy, the level of SFB was significantly higher in the SFB group than in the CON group, while a significant decrease in total Enterobacteriaceae and Salmonella was observed in the ceca of the SFB group. RT-qPCR revealed increased expression of ß-defensin 14, and cytokines IL-10 and IFNγ. Discussion: The introduction of SFB at hatch as a prophylactic treatment may benefit commercial partners as well as consumers by reducing the incidence of Enterobacteriaceae in food animals. Reduction of these bacteria in animals would, in turn, increase animal health, productivity, and safety for consumers. Studies to optimize the treatment for poultry industry applications are ongoing in our lab.

10.
Front Microbiol ; 13: 928346, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35875575

RESUMO

Bovine mastitis is an inflammation of the udder tissue parenchyma that causes pathological changes in the glandular tissue and abnormalities in milk leading to significant economic losses to the dairy industry across the world. Mammary pathogenic Escherichia (E.) coli (MPEC) is one of the main etiologic agents of acute clinical mastitis in dairy cattle. MPEC strains have virulence attributes to resist the host innate defenses and thrive in the mammary gland environment. The association between specific virulence factors of MPEC with the severity of mastitis in cattle is not fully understood. Furthermore, the indiscriminate use of antibiotics to treat mastitis has resulted in antimicrobial resistance to all major antibiotic classes in MPEC. A thorough understanding of MPEC's pathogenesis and antimicrobial susceptibility pattern is required to develop better interventions to reduce mastitis incidence and prevalence in cattle and the environment. This review compiles important information on mastitis caused by MPEC (e.g., types of mastitis, host immune response, diagnosis, treatment, and control of the disease) as well as the current knowledge on MPEC virulence factors, antimicrobial resistance, and the dilemma of MPEC as a new pathotype. The information provided in this review is critical to identifying gaps in knowledge that will guide future studies to better design diagnostic, prevent, and develop therapeutic interventions for this significant dairy disease.

11.
Front Microbiol ; 13: 891548, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35847067

RESUMO

The emergence of new antimicrobial resistant and virulent bacterial strains may pose a threat to human and animal health. Bacterial plasmid conjugation is a significant contributor to rapid microbial evolutions that results in the emergence and spread of antimicrobial resistance (AR). The gut of animals is believed to be a potent reservoir for the spread of AR and virulence genes through the horizontal exchange of mobile genetic elements such as plasmids. The study of the plasmid transfer process in the complex gut environment is limited due to the confounding factors that affect colonization, persistence, and plasmid conjugation. Furthermore, study of plasmid transfer in the gut of humans is limited to observational studies, leading to the need to identify alternate models that provide insight into the factors regulating conjugation in the gut. This review discusses key studies on the current models for in silico, in vitro, and in vivo modeling of bacterial conjugation, and their ability to reflect the gut of animals. We particularly emphasize the use of computational and in vitro models that may approximate aspects of the gut, as well as animal models that represent in vivo conditions to a greater extent. Directions on future research studies in the field are provided.

12.
mSphere ; 6(5): e0069821, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34668756

RESUMO

Horizontal gene transfer (HGT) is a driving force of microbial evolution. The gut of animals acts as a potent reservoir for the lateral transfer of virulence, fitness, and antimicrobial resistance genes through plasmids. Reduced-complexity models for the examination of host-microbe interactions involved in plasmid transfer are greatly desired. Thus, this study identifies the use of Drosophila melanogaster as a model organism for the conjugation of plasmids of various incompatibility groups in the gut. Enterobacteriaceae conjugation pairs were identified in vitro and used for oral inoculation of the Drosophila gut. Flies were enumerated for the donor, recipient, and transconjugant populations. Each donor-recipient pair was observed to persist in fly guts for the duration of the experiment. Gut concentrations of the donors and recipients were significantly different between male and female flies, with females generally demonstrating increased concentrations. Furthermore, host genetics significantly altered the concentrations of donors and recipients. However, transconjugant concentrations were not affected by host sex or genetics and were detected only in the IncPε and IncI1 plasmid groups. This study demonstrates Drosophila melanogaster as a model for gut-mediated plasmid transfer. IMPORTANCE Microbial evolution in the gut of animals due to horizontal gene transfer (HGT) is of significant interest for microbial evolution as well as within the context of human and animal health. Microbial populations evolve within the host, and factors from the bacteria and host interact to regulate this evolution. However, little is currently known about how host and bacterial factors regulate plasmid-mediated HGT in the gut. This study demonstrates the use of Drosophila and the roles of sexual dimorphism as well as plasmid incompatibility groups in HGT in the gut.


Assuntos
Drosophila melanogaster/microbiologia , Farmacorresistência Bacteriana Múltipla/genética , Microbioma Gastrointestinal/genética , Transferência Genética Horizontal/genética , Plasmídeos/genética , Animais , Bactérias/genética , Bactérias/isolamento & purificação , Feminino , Especificidade de Hospedeiro , Masculino , Caracteres Sexuais , Virulência
13.
Commun Biol ; 4(1): 1359, 2021 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-34862463

RESUMO

Salmonella enterica persist in the chicken gut by suppressing inflammatory responses via expansion of intestinal regulatory T cells (Tregs). In humans, T cell activation is controlled by neurochemical signaling in Tregs; however, whether similar neuroimmunological signaling occurs in chickens is currently unknown. In this study, we explore the role of the neuroimmunological axis in intestinal Salmonella resistance using the drug reserpine, which disrupts intracellular storage of catecholamines like norepinephrine. Following reserpine treatment, norepinephrine release was increased in both ceca explant media and Tregs. Similarly, Salmonella killing was greater in reserpine-treated explants, and oral reserpine treatment reduced the level of intestinal Salmonella Typhimurium and other Enterobacteriaceae in vivo. These antimicrobial responses were linked to an increase in antimicrobial peptide and IL-2 gene expression as well as a decrease in CTLA-4 gene expression. Globally, reserpine treatment led to phosphorylative changes in epidermal growth factor receptor (EGFR), mammalian target of rapamycin (mTOR), and the mitogen-associated protein kinase 2(MEK2). Exogenous norepinephrine treatment alone increased Salmonella resistance, and reserpine-induced antimicrobial responses were blocked using beta-adrenergic receptor inhibitors, suggesting norepinephrine signaling is crucial in this mechanism. Furthermore, EGF treatment reversed reserpine-induced antimicrobial responses, whereas mTOR inhibition increased antimicrobial activities, confirming the roles of metabolic signaling in these responses. Finally, MEK1/2 inhibition suppressed reserpine, norepinephrine, and mTOR-induced antimicrobial responses. Overall, this study demonstrates a central role for MEK1/2 activity in reserpine induced neuro-immunometabolic signaling and subsequent antimicrobial responses in the chicken intestine, providing a means of reducing bacterial colonization in chickens to improve food safety.


Assuntos
Galinhas , Resistência à Doença/efeitos dos fármacos , Infecções por Enterobacteriaceae/veterinária , Enterobacteriaceae/fisiologia , Doenças das Aves Domésticas/microbiologia , Reserpina/farmacologia , Transdução de Sinais , Animais , Infecções por Enterobacteriaceae/microbiologia , Intestinos/imunologia , Intestinos/microbiologia , Salmonelose Animal/microbiologia , Salmonella typhimurium/fisiologia
14.
Microorganisms ; 9(1)2021 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-33435450

RESUMO

Resistance to last resort antibiotics in bacteria is an emerging threat to human and animal health. It is important to identify the source of these antimicrobial resistant (AMR) bacteria that are resistant to clinically important antibiotics and evaluate their potential transfer among bacteria. The objectives of this study were to (i) detect bacteria resistant to colistin, carbapenems, and ß-lactams in commercial poultry farms, (ii) characterize phylogenetic and virulence markers of E. coli isolates to potentiate virulence risk, and (iii) assess potential transfer of AMR from these isolates via conjugation. Ceca contents from laying hens from conventional cage (CC) and cage-free (CF) farms at three maturity stages were randomly sampled and screened for extended-spectrum ß-lactamase (ESBL)-producing Enterobacteriaceae, carbapenem-resistant Acinetobacter (CRA), and colistin resistant Escherichia coli (CRE) using CHROMagar™ selective media. We found a wide-spread abundance of CRE in both CC and CF hens across all three maturity stages. Extraintestinal pathogenic Escherichia coli phylogenetic groups B2 and D, as well as plasmidic virulence markers iss and iutA, were widely associated with AMR E. coli isolates. ESBL-producing Enterobacteriaceae were uniquely detected in the early lay period of both CC and CF, while multidrug resistant (MDR) Acinetobacter were found in peak and late lay periods of both CC and CF. CRA was detected in CF hens only. blaCMY was detected in ESBL-producing E. coli in CC and CF and MDR Acinetobacter spp. in CC. Finally, the blaCMY was shown to be transferrable via an IncK/B plasmid in CC. The presence of MDR to the last-resort antibiotics that are transferable between bacteria in food-producing animals is alarming and warrants studies to develop strategies for their mitigation in the environment.

15.
Infect Immun ; 78(4): 1528-41, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20086082

RESUMO

Despite the fact that the presence of multiple large plasmids is a defining feature of extraintestinal pathogenic Escherichia coli (ExPEC), such as avian pathogenic E. coli (APEC), and despite the fact that these bacteria pose a considerable threat to both human and animal health, characterization of these plasmids is still limited. In this study, after successfully curing APEC of its plasmids, we were able to investigate, for the first time, the contribution to virulence of three plasmids, pAPEC-1 (103 kb), pAPEC-2 (90 kb), and pAPEC-3 (60 kb), from APEC strain chi7122 individually as well as in all combinations in the wild-type background. Characterization of the different strains revealed unique features of APEC virulence. In vivo assays showed that curing the three plasmids resulted in severe attenuation of virulence. The presence of different plasmids and combinations of plasmids resulted in strains with different pathotypes and levels of virulence, reflecting the diversity of APEC strains associated with colibacillosis in chickens. Unexpectedly, our results associated the decrease in growth of some strains in some media with the virulence of APEC, and the mechanism was associated with some combinations of plasmids that included pAPEC-1. This study provided new insights into the roles of large plasmids in the virulence, growth, and evolution of APEC by showing for the first time that both the nature of plasmids and combinations of plasmids have an effect on these phenomena. It also provided a plausible explanation for some of the conflicting results related to the virulence of ExPEC strains. This study should help us understand the virulence of other ExPEC strains and design more efficient infection control strategies.


Assuntos
Infecções por Escherichia coli/veterinária , Proteínas de Escherichia coli/genética , Escherichia coli/patogenicidade , Plasmídeos , Doenças das Aves Domésticas/microbiologia , Fatores de Virulência/genética , Animais , Galinhas , DNA Bacteriano/genética , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/isolamento & purificação , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/mortalidade , Genótipo , Dados de Sequência Molecular , Doenças das Aves Domésticas/mortalidade , Análise de Sequência de DNA , Análise de Sobrevida , Virulência
16.
Microorganisms ; 8(11)2020 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-33120988

RESUMO

Foodborne pathogens are a public health threat globally. Shiga toxin-producing Escherichia coli (STEC), particularly O26, O111, and O157 STEC, are often associated with foodborne illness in humans. To create effective preharvest interventions, it is critical to understand which factors STEC strains use to colonize the gastrointestinal tract of cattle, which serves as the reservoir for these pathogens. Several colonization factors are known, but little is understood about initial STEC colonization factors. Our objective was to identify these factors via contrasting gene expression between nonpathogenic E. coli and STEC. Colonic explants were inoculated with nonpathogenic E. coli strain MG1655 or STEC strains (O26, O111, or O157), bacterial colonization levels were determined, and RNA was isolated and sequenced. STEC strains adhered to colonic explants at numerically but not significantly higher levels compared to MG1655. After incubation with colonic explants, flagellin (fliC) was upregulated (log2 fold-change = 4.0, p < 0.0001) in O157 STEC, and collectively, Lon protease (lon) was upregulated (log2 fold-change = 3.6, p = 0.0009) in STEC strains compared to MG1655. These results demonstrate that H7 flagellum and Lon protease may play roles in early colonization and could be potential targets to reduce colonization in cattle.

17.
Front Vet Sci ; 7: 592312, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33195630

RESUMO

Commercial poultry farms frequently use live bacterial prophylactics like vaccines and probiotics to prevent bacterial infections. Due to the emergence of antibiotic-resistant bacteria in poultry animals, a closer examination into the health benefits and limitations of commercial, live prophylactics as an alternative to antibiotics is urgently needed. In this review, we summarize the peer-reviewed literature of several commercial live bacterial vaccines and probiotics. Per our estimation, there is a paucity of peer-reviewed published research regarding these products, making repeatability, product-comparison, and understanding biological mechanisms difficult. Furthermore, we briefly-outline significant issues such as probiotic-label accuracy, lack of commercially available live bacterial vaccines for major poultry-related bacteria such as Campylobacter and Clostridium perfringens, as well research gaps (i.e., probiotic-mediated vaccine adjuvancy, gut-brain-microbiota axis). Increased emphasis on these areas would open several avenues for research, ranging from improving protection against bacterial pathogens to using these prophylactics to modulate animal behavior.

18.
Front Vet Sci ; 7: 629, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33102558

RESUMO

The animal gut is a major site affecting productivity via its role in mediating functions like food conversion and pathogen colonization. Live microorganisms like probiotics are widely used to improve poultry productivity. However, given that chicks receive their microbiota from the environment at-hatch, a bacterial treatment that can stimulate gut immune maturation in early life can benefit animal health. Thus, our lab has begun investigating alternative means to improve poultry health via single inoculation with microbial spores. In this study, we orally-inoculated day-old chicks with ileal scrapings (ISs) enriched for spores via chloroform treatment (SPORE) or non-treated (CON). At 3, 7, and 14 days post-inoculation (dpi), gut permeability was measured via FITC-dextran assay in serum. Additionally, small intestinal scrapings (SISs) were tested for in vitro Salmonella killing and total IgA. Lastly, distal ileum was either fixed or flash-frozen for microscopy or kinome peptide array, respectively. Using bacterial 16S rRNA gene sequencing, SPORE and CON inocula were highly-similar in bacterial composition. However, spores were detected in SPORE but not in CON inoculum. Segmented filamentous bacteria (SFB) filaments were observed in the distal ileum in SPORE birds as early as 3 dpi and all birds at 7 and 14 dpi. Additionally, SFB were detected via PCR in the ceca, colonizing all SPORE birds at 3 dpi. At 3 dpi, SPORE birds exhibited lower gut permeability vs. CON. In SPORE birds, SISs induced greater Salmonella growth in vitro at 3 dpi yet significantly-reduced Salmonella load at 7 and 14 dpi compared to CON in an IgA-independent manner. SPORE distal ileal tissue exhibited unique upregulation of several immunometabolic processes vs. CON birds, including innate (Toll-like receptor, JAK-STAT) and adaptive (T/B cell receptor, TH17 differentiation) immune pathways, PI3K/Akt signaling, mTOR signaling, and insulin-related pathways. Collectively, these data suggest oral inoculation with ileal spores generally-improved gut health. Importance: We report that ileal, spore-forming commensal microbes have potent effects on ileum immunometabolism. Additionally, we identify a functional ileal phenotype in spore-treated chickens, which matched several of the observed immunometabolic changes and was associated with SFB colonization in the ileum.

19.
Front Microbiol ; 11: 625286, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33519786

RESUMO

Chicken intestinal Escherichia coli are a reservoir for virulence and antimicrobial resistance (AMR) genes that are often carried on incompatibility group F (IncF) plasmids. The rapid transfer of these plasmids between bacteria in the gut contributes to the emergence of new multidrug-resistant and virulent bacteria that threaten animal agriculture and human health. Thus, the aim of the present study was to determine whether live bacterial prophylactics could affect the distribution of large virulence plasmids and AMR in the intestinal tract and the potential role of smRNA in this process. In this study, we tested ∼100 randomly selected E. coli from pullet feces (n = 3 per group) given no treatment (CON), probiotics (PRO), a live Salmonella vaccine (VAX), or both (P + V). E. coli isolates were evaluated via plasmid profiles and several phenotypic (siderophore production and AMR), and genotypic (PCR for virulence genes and plasmid typing) screens. P + V isolates exhibited markedly attenuated siderophore production, lack of AMR and virulence genes, which are all related to the loss of IncF and ColV plasmids (P < 0.0001). To identify a causal mechanism, we evaluated smRNA levels in the ceca mucus and found a positive association between smRNA concentrations and plasmid content, with both being significantly reduced in P + V birds compared to other groups (P < 0.01). To test this positive association between IncF plasmid transfer and host smRNA concentration, we evenly pooled smRNA per group and treated E. coli mating pairs with serial concentrations of smRNA in vitro. Higher smRNA concentrations resulted in greater rates of IncF plasmid transfer between E. coli donors (APEC O2 or VAX isolate IA-EC-001) and recipient (HS-4) (all groups; P < 0.05). Finally, RNAHybrid predictive analyses detected several chicken miRNAs that hybridize with pilus assembly and plasmid transfer genes on the IncF plasmid pAPEC-O2-R. Overall, we demonstrated P + V treatment reduced smRNA levels in the chicken ceca, which was associated with a reduction in potentially virulent E. coli. Furthermore, we propose a novel mechanism in which intestinal smRNAs signal plasmid exchange between E. coli. Investigations to understand the changes in bacterial gene expression as well as smRNAs responsible for this phenomenon are currently underway.

20.
Poult Sci ; 99(2): 752-762, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32029160

RESUMO

Commercial poultry farms are increasingly threatened by bacterial infections from avian pathogenic Escherichia coli (APEC) and broad-host Salmonella serovars. Recombinant attenuated Salmonella vaccines (RASV) elicit cross-reactive immune responses against APEC in chickens; however, assessment of broad protection is lacking. Probiotics boost chicken immunity and improve vaccination responses. The objective of this study was to determine whether the RASV, the probiotics, or their combination had protection against APEC and Salmonella. White Leghorn chicks were randomly placed into 4 groups: no treatment (CON), probiotics (PRO), RASV (VAX), or both prophylactics (P + V). Chicks in the PRO and P + V groups were fed probiotics daily, beginning at the age of 1-day-old. Chicks in the P + V and VAX groups were orally inoculated with RASV at the age of 4 D and boosted 2 wks later. Total and antigen-specific IgY responses to Salmonella (lipolysaccharide [LPS]) and E. coli (IroN and IutA) were measured in serum samples via ELISA. Bactericidal potential of both serum and blood against 42 APEC isolates comprising 25 serotypes was assessed in vitro. In vivo protection against APEC was evaluated by air sac challenge with APEC χ7122 (O78:K80), gross pathological lesions were scored, and bacterial loads were enumerated. In a second similar study, birds were orally challenged with S. Kentucky (CVM29188), and feces were enumerated for Salmonella at multiple time points. Vaccination elicited significant LPS-specific antibodies regardless of probiotics (P < 0.0001). Chicks in the P + V group demonstrated increased blood and serum bactericidal abilities against multiple APEC strains in vitro compared with the CON group. Following χ7122 challenge, P+V birds had less APEC in their blood (P < 0.001) and lower signs of airsacculitis (P < 0.01) and pericarditis/perihepatitis (P < 0.05) than CON birds. Finally, only P + V birds were negative for fecal Salmonella at all time points. This study shows this combination treatment may be a feasible method to reduce infection by APEC and Salmonella in chickens.


Assuntos
Galinhas , Infecções por Escherichia coli/veterinária , Escherichia coli/efeitos dos fármacos , Doenças das Aves Domésticas/terapia , Probióticos/farmacologia , Salmonelose Animal/terapia , Vacinas contra Salmonella/imunologia , Salmonella enterica/imunologia , Animais , Infecções por Escherichia coli/terapia , Feminino , Masculino , Organismos Livres de Patógenos Específicos , Vacinas Atenuadas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA