Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Vet Microbiol ; 259: 109135, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34090248

RESUMO

Bovine respiratory disease (BRD) is caused by a mixture of viruses and opportunistic bacteria belonging to Pasteurellaceae and Mycoplasma bovis. However, these organisms are also commonly isolated from healthy calves. This study aimed to determine whether the organisms are present in higher numbers in calves sick with acute BRD than in clinically healthy calves, and further to genetically characterize bacteria of the family Pasteurellaceae to understand whether particular types are associated with disease. Forty-six clinically healthy and 46 calves with BRD were sampled by broncheoalveolar lavage (BAL) method in 11 herds geographically spread over Denmark to determine presence and quantity of microorganisms by culture and quantitative real time qPCR. Isolates of Pasteurellaceae were tested for antibiotic resistance and were whole genome sequenced to determine genotypes. Histophilus somni was in particular positively associated with BRD, suggesting particular importance of this organism as likely aetiology of BRD. In addition, quantification of bacteria revealed that higher counts of H. somni as well as of M. haemolytica was also a good indicator of the disease. Pasteurellaceae isolates were susceptible to the commonly used antibiotics in treatment of BRD, and genotypes were shared between isolates from clinically healthy and sick calves.


Assuntos
Bactérias/genética , Bactérias/patogenicidade , Complexo Respiratório Bovino/microbiologia , Doenças dos Bovinos/virologia , Doenças Respiratórias/microbiologia , Doenças Respiratórias/veterinária , Animais , Antibacterianos/farmacologia , Bactérias/classificação , Bactérias/isolamento & purificação , Líquido da Lavagem Broncoalveolar/microbiologia , Líquido da Lavagem Broncoalveolar/virologia , Bovinos , Mannheimia haemolytica/genética , Mannheimia haemolytica/isolamento & purificação , Mannheimia haemolytica/patogenicidade , Pasteurellaceae/classificação , Pasteurellaceae/efeitos dos fármacos , Pasteurellaceae/genética , Pasteurellaceae/patogenicidade , Doenças Respiratórias/virologia
2.
Molecules ; 26(5)2021 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-33652818

RESUMO

Glaesserella parasuis (G. parasuis) causes inflammation and damage to piglets. Whether polyserositis caused by G. parasuis is due to tight junctions damage and the protective effect of baicalin on it have not been examined. Therefore, this study aims to investigate the effects of baicalin on peritoneal tight junctions of piglets challenged with G. parasuis and its underlying molecular mechanisms. Piglets were challenged with G. parasuis and treated with or without baicalin. RT-PCR was performed to examine the expression of peritoneal tight junctions genes. Immunofluorescence was carried out to detect the distribution patterns of tight junctions proteins. Western blot assays were carried out to determine the involved signaling pathways. Our data showed that G. parasuis infection can down-regulate the tight junctions expression and disrupt the distribution of tight junctions proteins. Baicalin can alleviate the down-regulation of tight junctions mRNA in peritoneum, prevent the abnormalities and maintain the continuous organization of tight junctions. Our results provide novel evidence to support that baicalin has the capacity to protect peritoneal tight junctions from G. parasuis-induced inflammation. The protective mechanisms of baicalin could be associated with inhibition of the activation of PKC and MLCK/MLC signaling pathway. Taken together, these data demonstrated that baicalin is a promising natural agent for the prevention and treatment of G. parasuis infection.


Assuntos
Flavonoides/farmacologia , Infecções por Pasteurellaceae/tratamento farmacológico , Pasteurellaceae/efeitos dos fármacos , Doenças dos Suínos/tratamento farmacológico , Animais , Pasteurellaceae/genética , Pasteurellaceae/patogenicidade , Infecções por Pasteurellaceae/genética , Infecções por Pasteurellaceae/microbiologia , Infecções por Pasteurellaceae/veterinária , Peritônio/efeitos dos fármacos , Peritônio/microbiologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética , Suínos , Doenças dos Suínos/microbiologia , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/genética , Junções Íntimas/microbiologia
3.
Microbes Infect ; 23(2-3): 104771, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33164813

RESUMO

Rodentibacter (R.) heylii is frequently detected in laboratory rodents. Repeats in toxin (RTX) toxins are considered important virulence factors of this major murine pathogen. We evaluated the virulence of a R.heylii strain negative for all known RTX toxin genes and Muribacter (M.) muris, a commensal in mice, in experimental infections of C57BL/6 and BALB/c mice. Experimental intranasal infection with 108 CFU of the pnxI-, pnxII- and pnxIII- R. heylii strain resulted in 75% and 100% mortality in C57BL/6 and BALB/c mice, respectively. In early losses, multiple internal organs were infected and purulent bronchopneumonia was the main pathology. Intranasal application of M. muris did not result in mortality or severe weight loss. Immunoproteomics led to the identification of a surface-associated and specific immunogen, which was designated as R. heylii immunogen A (RhiA) and which was exclusively recognised by sera obtained from mice infected with this R. heylii pathotype. RhiA is a 262.6 kDa large protein containing long imperfect tandem repeats and C-terminal RTX consensus sequences. Immunohistochemical analysis confirmed that this R.heylii pathotype expresses RhiA in the lower respiratory tract. In summary, this study describes a specific immunogen in a virulent R. heylii, strain which is an excellent antigen for pathotype-specific serological screenings and which might carry out RTX-related functions.


Assuntos
Proteínas de Bactérias/imunologia , Infecções por Pasteurellaceae/veterinária , Pasteurellaceae/imunologia , Doenças dos Roedores/microbiologia , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Sequência Consenso , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pasteurellaceae/química , Pasteurellaceae/genética , Pasteurellaceae/patogenicidade , Infecções por Pasteurellaceae/microbiologia , Infecções por Pasteurellaceae/mortalidade , Domínios Proteicos , Doenças dos Roedores/mortalidade , Sequências de Repetição em Tandem , Virulência , Fatores de Virulência/química , Fatores de Virulência/genética , Fatores de Virulência/imunologia
4.
Infect Immun ; 89(2)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33139386

RESUMO

S-Ribosylhomocysteinase (LuxS) is required for the synthesis of the autoinducer-2 (AI-2) quorum-sensing signaling molecule in many Gram-negative bacteria. The bovine (and ovine) opportunistic pathogen Histophilus somni contains luxS and forms a biofilm containing an exopolysaccharide (EPS) in the matrix. Since biofilm formation is regulated by quorum sensing in many bacteria, the roles of luxS in H. somni virulence and biofilm formation were investigated. Although culture supernatants from H. somni were ineffective at inducing bioluminescence in the Vibrio harveyi reporter strain BB170, H. somniluxS complemented the biosynthesis of AI-2 in the luxS-deficient Escherichia coli strain DH5α. H. somni strain 2336 luxS was inactivated by transposon mutagenesis. RNA expression profiles revealed that many genes were significantly differentially expressed in the luxS mutant compared to that in the wild-type, whether the bacteria were grown planktonically or in a biofilm. Furthermore, the luxS mutant had a truncated and asialylated lipooligosaccharide (LOS) and was substantially more serum sensitive than the wild-type. Not surprisingly, the luxS mutant was attenuated in a mouse model for H. somni virulence, and some of the altered phenotypes were partially restored after the mutation was complemented with a functional luxS However, no major differences were observed between the wild-type and the luxS mutant in regard to outer membrane protein profiles, biofilm formation, EPS production, or intracellular survival. These results indicate that luxS plays a role in H. somni virulence in the context of LOS biosynthesis but not biofilm formation or other phenotypic properties examined.


Assuntos
Proteínas de Bactérias/imunologia , Liases de Carbono-Enxofre/imunologia , Lipopolissacarídeos/biossíntese , Lipopolissacarídeos/imunologia , Infecções por Pasteurellaceae/imunologia , Pasteurellaceae/genética , Pasteurellaceae/imunologia , Pasteurellaceae/patogenicidade , Virulência/imunologia , Animais , Proteínas de Bactérias/genética , Biofilmes , Liases de Carbono-Enxofre/genética , Bovinos , Modelos Animais de Doenças , Variação Genética , Genótipo , Humanos , Camundongos , Infecções por Pasteurellaceae/genética , Percepção de Quorum/imunologia , Ovinos
5.
Mol Cell Probes ; 53: 101641, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32735874

RESUMO

Gallibacterium anatis (G. anatis), one of the major pathogens causing reproductive tract disorders in laying hens, leads to a reduction in egg production and increased mortality, caused by either single or mixed infections with other pathogens. As a specific virulence factor of G. anatis, the role of GtxA in layers' salpingitis remains unclear. In this study, we explored the effect of GtxA on G. anatis infection by comparing wild strain Yu-PDS-RZ-1-SLG (RZ) and its GtxA deleted counterpart RZΔgtxA in primary chicken oviduct epithelial cells (COEC). Their adherence, invasion, cytoxicity, and ability to induce apoptosis and and cytokine secretion were evaluated and the cytotoxicity and cytokine secretion of the recombinant GtxA protein and its N-terminal adenylate cyclase and C-terminal RTX hemolysin domain were also analyzed. We found that the adhesion ability of RZΔgtxA was significantly lower than that of parental strain RZ, and its toxicity to COEC was weakened; Meanwhile, apoptosis was inhibited and the expression of IL-6, IL-2, TNF-α and IFN-γ were dramatically reduced in COEC infected by RZΔgtxA. In contrast, the recombinant protein GtxA inhibited the proliferation of oviduct cells and induced obvious cytotoxicity, and the expression of IL-6, TNF-α and IFN-γ were up-regulated in COEC interacted with recombinant proteins. Our study indicates that GtxA promotes G. anatis adherence to cells, changes cells permeability and expression of inflammatory factors, resulting in cell damage and apoptosis.


Assuntos
Toxinas Bacterianas/genética , Infecções por Pasteurellaceae/veterinária , Pasteurellaceae/patogenicidade , Doenças das Aves Domésticas/microbiologia , Animais , Aderência Bacteriana , Galinhas , Citocinas/metabolismo , Células Epiteliais/citologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Feminino , Deleção de Genes , Oviductos/citologia , Oviductos/imunologia , Oviductos/microbiologia , Pasteurellaceae/genética , Pasteurellaceae/imunologia , Infecções por Pasteurellaceae/imunologia , Fatores de Virulência/genética
6.
Vet Res ; 51(1): 40, 2020 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-32156313

RESUMO

GtxA, a leukotoxic RTX-toxin, has been proposed a main virulence factor of Gallibacterium anatis. To evaluate the impact of GtxA during infection, we experimentally infected laying hens with a G. anatis wild-type (WT) strain and its isogenic gtxA deletion mutant (ΔgtxA), respectively, and monitored the birds during a 6 day period. Birds inoculated with ΔgtxA had significantly reduced gross lesions and microscopic changes compared to the birds inoculated with the WT strain. To assess the host response further, we quantified the expression of pro-inflammatory cytokines and apoptosis genes by RT-qPCR. In the ovarian tissue, the expression levels of IL-4 and TNF-α were significantly lower in the ΔgtxA group compared to the WT group, while IL-6 and IL-10 levels appeared similar in the two groups. In the spleen tissue of ΔgtxA infected chickens, IL-4 expression was also lower compared to the WT infected chickens. The results indicated that GtxA plays a key role in an acute cytokine-mediated Th2-like response against G. anatis infection in the ovary tissue. The pro-inflammatory response in the ovary tissue of birds inoculated with ΔgtxA mutant was thus significantly lower than the wild-type response. This was, at least partly, supported by the apoptosis gene expression levels, which were significantly higher in the ΔgtxA mutant compared to the wild-type infected chickens. In conclusion, GtxA clearly plays an important role in the pathogenesis of G. anatis infections in laying hens. Further investigations into the specific factors regulating the host response is however needed to provide a more complete understanding of the bacteria-host interaction.


Assuntos
Proteínas de Bactérias/genética , Galinhas , Infecções por Pasteurellaceae/veterinária , Pasteurellaceae/patogenicidade , Doenças das Aves Domésticas/microbiologia , Fatores de Virulência/genética , Animais , Proteínas de Bactérias/metabolismo , Feminino , Pasteurellaceae/genética , Pasteurellaceae/fisiologia , Infecções por Pasteurellaceae/microbiologia , Fatores de Virulência/metabolismo
7.
Vet Q ; 40(1): 16-34, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31902298

RESUMO

Gallibacterium anatis is a Gram-negative bacterium of the Pasteurellaceae family that resides normally in the respiratory and reproductive tracts in poultry. It is a major cause of oophoritis, salpingitis, and peritonitis, decreases egg production and mortality in hens thereby severely affecting animal welfare and overall productivity by poultry industries across Europe, Asia, America, and Africa. In addition, it has the ability to infect wider host range including domesticated and free-ranging avian hosts as well as mammalian hosts such as cattle, pigs and human. Evaluating the common virulence factors including outer membrane vesicles, fimbriae, capsule, metalloproteases, biofilm formation, hemagglutinin, and determining novel factors such as the RTX-like toxin GtxA, elongation factor-Tu, and clustered regularly interspaced short palindromic repeats (CRISPR) has pathobiological, diagnostic, prophylactic, and therapeutic significance. Treating this bacterial pathogen with traditional antimicrobial drugs is discouraged owing to the emergence of widespread multidrug resistance, whereas the efficacy of preventing this disease by classical vaccines is limited due to its antigenic diversity. It will be necessary to acquire in-depth knowledge on important virulence factors, pathogenesis and, concerns of rising antibiotic resistance, improvised treatment regimes, and novel vaccine candidates to effectively tackle this pathogen. This review substantially describes the etio-epidemiological aspects of G. anatis infection in poultry, and updates the recent development in understanding the pathogenesis, organism evolution and therapeutic and prophylactic approaches to counter G. anatis infection for safeguarding the welfare and health of poultry.


Assuntos
Infecções por Pasteurellaceae/veterinária , Doenças das Aves Domésticas , Animais , Antibacterianos/uso terapêutico , Vacinas Bacterianas , Pasteurellaceae/imunologia , Pasteurellaceae/patogenicidade , Infecções por Pasteurellaceae/diagnóstico , Infecções por Pasteurellaceae/tratamento farmacológico , Infecções por Pasteurellaceae/epidemiologia , Aves Domésticas , Doenças das Aves Domésticas/diagnóstico , Doenças das Aves Domésticas/tratamento farmacológico , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/patologia
8.
Avian Dis ; 64(4): 536-541, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33570106

RESUMO

The prevalence of Gallibacterium anatis in poultry production has increased over the last two decades. However, only a few studies have explored the pathogenicity of this bacterium in commercial layer chickens. This trial studied the aspects of the pathogenicity of a Gallibacterium anatis biovar haemolytica local Egyptian isolate (previously registered as strain B14 with GenBank accession no. KJ026147). We used 500 base pairs of a 16S ribosomal RNA gene and the 16S-23S ribosomal RNA intergenic spacer, partial sequence in an experimental infection trial in commercial White Shaver layer chickens aged 19 wk. The hens were divided into three groups of 40 birds each. The hens in Groups 1 and 2 were experimentally infected through the intranasal (IN) and intravenous (IV) routes, respectively, with a dose of 0.2 ml/bird containing 1.2 × 109 colony-forming units/ml. In contrast, Group 3 was kept as a noninfected control group. Both IN and IV infections resulted in a delayed egg laying for 1 wk and a significant (P ≤ 0.05) drop in egg production by 7.81% and 10.28% compared with the control group over 7 wk. Severe lesions in the form of hemorrhagic pneumonia, catarrhal tracheitis, ovarian follicle and oviductal regression, and septicemia were evident on necropsy, demonstrating the pathogenicity of G. anatis as a primary pathogen.


Assuntos
Galinhas , Doenças Ovarianas/veterinária , Infecções por Pasteurellaceae/veterinária , Pasteurellaceae/fisiologia , Pasteurellaceae/patogenicidade , Doenças das Aves Domésticas/patologia , Doenças Respiratórias/veterinária , Animais , Feminino , Doenças Ovarianas/microbiologia , Doenças Ovarianas/patologia , Doenças Ovarianas/fisiopatologia , Pasteurellaceae/genética , Infecções por Pasteurellaceae/microbiologia , Infecções por Pasteurellaceae/patologia , Infecções por Pasteurellaceae/fisiopatologia , Doenças das Aves Domésticas/microbiologia , Doenças das Aves Domésticas/fisiopatologia , Doenças Respiratórias/microbiologia , Doenças Respiratórias/patologia , Doenças Respiratórias/fisiopatologia , Sepse/microbiologia , Sepse/patologia , Sepse/fisiopatologia , Sepse/veterinária
9.
Avian Pathol ; 48(6): 549-556, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31280592

RESUMO

This study reports the results of diagnostic and molecular typing methods for 18 Avibacterium paragallinarum isolates obtained from outbreaks of infectious coryza in commercial layer flocks in the Netherlands. Isolation, biochemical identification, species-specific PCR tests and classical serotyping were performed. In addition, molecular typing by Enterobacterial Repetitive Intergenic Consensus-Based Polymerase Chain Reaction (ERIC-PCR) and sequence analysis of the partial HPG2 region of A. paragallinarum were applied and results of both techniques were compared. Moreover, the pathogenicity of an isolate of the most common genotype detected in the Netherlands was determined in an animal experiment. All 18 Avibacterium isolates were nicotinamide adenine dinucleotide-dependent. All isolates were detected by the species-specific conventional PCR while 33% of the isolates were missed by the species-specific real-time PCR. Sequence analysis showed a probe mismatch as a result of a single nucleotide polymorphism (G1516A). Modification of the probe of the real-time PCR was necessary to overcome false negative results. Molecular typing showed that sequence analysis of the partial HPG2 region was in concordance with ERIC-PCR results and indicated the presence of two major genotypes. Serotyping showed the presence of serovars A-1, A-2 and B-1. There was no correlation between genotyping results and serotyping results. Inoculation of an isolate of the most prevalent genotype, and belonging to serovar A-1, into brown layer hens demonstrated the pathogenicity of this isolate.


Assuntos
Galinhas/microbiologia , Enterobacteriaceae/genética , Infecções por Pasteurellaceae/veterinária , Pasteurellaceae/genética , Doenças das Aves Domésticas/microbiologia , Animais , Surtos de Doenças/veterinária , Enterobacteriaceae/isolamento & purificação , Feminino , Tipagem Molecular/veterinária , Países Baixos/epidemiologia , Pasteurellaceae/isolamento & purificação , Pasteurellaceae/patogenicidade , Infecções por Pasteurellaceae/epidemiologia , Infecções por Pasteurellaceae/microbiologia , Reação em Cadeia da Polimerase/veterinária , Doenças das Aves Domésticas/diagnóstico , Doenças das Aves Domésticas/epidemiologia , Sorogrupo , Sorotipagem/veterinária , Especificidade da Espécie , Virulência
10.
BMC Microbiol ; 19(1): 43, 2019 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-30777007

RESUMO

BACKGROUND: Rodentibacter (R.) pneumotropicus colonizes the respiratory and urogenital tracts of laboratory mice with a reported moderate serological prevalence from 4 to 13%. Thus, regular tests to identify this pathogen in mice are recommended for animal facilities. However, a recent study indicated that current serological assays are partly insensitive, as C57BL/6 and BALB/c mice infected with R. pneumotropicus were incorrectly screened as seronegative. RESULTS: Here, we report a systematic analysis of protein and lipopolysaccharides antigens by immunoblot and ELISA that allowed establishing a sensitive test system able to differentiate between R. pneumotropicus and the closely related species R. heylii. Furthermore, the main immunogen, designated as 'characteristic antigen for Rodentibacter of laboratory origin 1' (CARLO-1), was identified by two-dimensional gel electrophoresis followed by immunoblot and tandem mass spectrometry in a preparation of outer membrane proteins. An indirect ELISA relying on the recombinantly expressed protein provided high sensitivity, specificity, and selectivity. The corresponding carlo1 gene was highly conserved (> 97%) among 21 isolates of R. pneumotropicus and R. heylii. CONCLUSION: The newly identified protein CARLO-1 is well suited for the sensitive and specific serological detection of Rodentibacter infections in mice. Indirect differentiation of R. pneumotropicus and R. heylii infections may be possible using an ELISA based on a whole-cell antigen preparation. All four established ELISA systems using a whole-cell preparation, lipopolysaccharides, outer-membrane proteins and protein CARLO-1 as antigen, respectively, outperformed a commercial ELISA in terms of sensitivity.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Ensaio de Imunoadsorção Enzimática , Infecções por Pasteurellaceae/diagnóstico , Infecções por Pasteurellaceae/veterinária , Pasteurellaceae/imunologia , Animais , Feminino , Immunoblotting , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pasteurellaceae/patogenicidade , Infecções por Pasteurellaceae/imunologia , Sensibilidade e Especificidade
11.
Infect Immun ; 86(12)2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30201700

RESUMO

Histophilus somni is capable of intracellular survival within professional phagocytic cells, but the mechanism of survival is not understood. The Fic motif within the direct repeat (DR1)/DR2 domains of the IbpA fibrillary network protein of H. somni is cytotoxic to epithelial and phagocytic cells, which may interfere with the bactericidal activity of these cells. To determine the contribution of IbpA and Fic to resistance to host defenses, H. somni strains and mutants that lacked all or a region of ibpA (including the DR1/DR2 regions) were tested for survival in bovine monocytic cells and for serum susceptibility. An H. somni mutant lacking IbpA, but not the DR1/DR2 region within ibpA, was more susceptible to killing by antiserum than the parent, indicating that the entire protein was associated with serum resistance. H. somni strains expressing IbpA replicated in bovine monocytes for at least 72 h and were toxic for these cells. Virulent strain 2336 mutants lacking the entire ibpA gene or both DR1 and DR2 were not toxic to the monocytes but still survived within the monocytes for at least 72 h. Monitoring of intracellular trafficking of H. somni with monoclonal antibodies to phagosomal markers indicated that the early phagosomal marker early endosome antigen 1 colocalized with all isolates tested, but only strains that could survive intracellularly did not colocalize with the late lysosomal marker lysosome-associated membrane protein 2 and prevented the acidification of phagosomes. These results indicated that virulent isolates of H. somni were capable of surviving within phagocytic cells through interference in phagosome-lysosome maturation. Therefore, H. somni may be considered a permissive intracellular pathogen.


Assuntos
Proteínas de Bactérias/imunologia , Lisossomos/metabolismo , Macrófagos/microbiologia , Pasteurellaceae/metabolismo , Fagossomos/metabolismo , Soro/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Proteínas de Bactérias/genética , Bovinos , Células Cultivadas , Lisossomos/microbiologia , Macrófagos/imunologia , Fusão de Membrana , Viabilidade Microbiana , Monócitos/microbiologia , Pasteurellaceae/patogenicidade , Fagocitose , Fagossomos/microbiologia
12.
PLoS One ; 13(6): e0198510, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29879182

RESUMO

Gfi1 is a key molecule in hematopoietic lineage development and mutations in GFI1 cause severe congenital neutropenia (SCN). Neutropenia is associated with low bone mass, but the underlying mechanisms are poorly characterized. Using Gfi1 knock-out mice (Gfi1-ko/ko) as SCN model, we studied the relationship between neutropenia and bone mass upon different pathogen load conditions. Our analysis reveals that Gfi1-ko/ko mice kept under strict specific pathogen free (SPF) conditions demonstrate normal bone mass and survival. However, Gfi1-ko/ko mice with early (nonSPF) or late (SPF+nonSPF) pathogen exposure develop low bone mass. Gfi1-ko/ko mice demonstrate a striking rise of systemic inflammatory markers according to elevated pathogen exposure and reduced bone mass. Elevated inflammatory cytokines include for instance Il-1b, Il-6, and Tnf-alpha that regulate osteoclast development. We conclude that low bone mass, due to low neutrophil counts, is caused by the degree of systemic inflammation promoting osteoclastogenesis.


Assuntos
Proteínas de Ligação a DNA/genética , Neutropenia/congênito , Osteoporose/etiologia , Fatores de Transcrição/genética , Animais , Peso Corporal , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/fisiologia , Diferenciação Celular , Síndrome Congênita de Insuficiência da Medula Óssea , Citocinas/genética , Citocinas/metabolismo , Proteínas de Ligação a DNA/deficiência , Extremidades/patologia , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutropenia/etiologia , Neutropenia/genética , Neutropenia/patologia , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese , Osteoporose/genética , Osteoporose/patologia , Osteoprotegerina/sangue , Pasteurellaceae/patogenicidade , Ligante RANK/sangue , Fatores de Transcrição/deficiência , Trichomonas/patogenicidade
13.
Vet Microbiol ; 217: 76-81, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29615261

RESUMO

In laying hens, Escherichia coli (E. coli) and Gallibacterium anatis (G. anatis) are considered the two major pathogens causing reproductive tract disorders, either as single infections or as co-infections. Vertical transmission has been confirmed for E. coli but remains to be clearly demonstrated for G. anatis. The aim of the present study was to investigate the ability of both G. anatis and E. coli at eggshell transmission using an embryonated egg dipping model, and to investigate the possible interaction between the two organisms in an embryonated egg injection model. Embryonated eggs were dipped into brain heart infusion broth containing 108 CFU/ml either of G. anatis 12656-12 liver, E. coli ST95 or E. coli ST141, respectively. E. coli ST95 and ST141 were re-isolated from the interior egg contents in 60% (12/20) and 85% (17/20) of the eggs, respectively, while G. anatis 12656-12 was only re-isolated from the interior egg contents in 6.7% (3/45) eggs. Eggs were injected with 10-1000 CFU of either G. anatis 12656-12, E. coli ST95 or ST141 into the allantoic cavity. As few as 10 CFU of G. anatis 12656-12 resulted in 100% mortality within 24 h post injection whereas the E. coli injected embryos all died at 48 h post injection. Significant difference in CFU counts were observed for G. anatis when compared G. anatis injection group with either of the two G. anatis - E. coli co-injection groups. Sixteen hours post injection, a significant difference in embryo mortality could be observed when comparing co-injected embryonated eggs (G. anatis and E. coli) and single-injected (G. anatis or E. coli) embryonated eggs. In conclusion, bacterial transmission via the eggshell was demonstrated for both G. anatis and E. coli although at different magnitudes. The embryonated egg injection model revealed that G. anatis in particular was highly pathogenic when exposed directly to the developing embryo.


Assuntos
Infecções por Escherichia coli/veterinária , Escherichia coli/patogenicidade , Óvulo/microbiologia , Infecções por Pasteurellaceae/veterinária , Pasteurellaceae/patogenicidade , Doenças das Aves Domésticas/transmissão , Animais , Embrião de Galinha , Galinhas/microbiologia , Infecções por Escherichia coli/transmissão , Feminino , Infecções por Pasteurellaceae/microbiologia , Infecções por Pasteurellaceae/transmissão , Doenças das Aves Domésticas/microbiologia , Virulência
14.
Vet Microbiol ; 203: 136-142, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28619135

RESUMO

Gallibacterium anatis (G. anatis) has been suggested to have a causal role in salpingitis and peritonitis in egg-laying chickens, leading to decreased egg production and increased mortality worldwide. Adherence and invasion of epithelial cells are thought to play a role in the pathogenesis of G. anatis infection. The purpose of this article was to study adherence and invasion of G. anatis using two G. anatis strains of different virulence (Yu-PDS-RZ-1-SLG strain, highly virulent and F149T strain, non-virulent) via infection of the primary chicken oviduct epithelial cells (PCOECs).The results showed that Yu-PDS-RZ-1 -SLG strain was able to attach to PCOECs at higher levels than that of F149T strain, but no invasion was observed with either strain. However, cell debris and cell apoptosis were observed after being exposed to G. anatis Yu-PDS-RZ-1-SLG for 90min, whereas G. anatis F149T did not cause cell damage, and adherence was prevented by trypsin treatment of bacterial cells. Cytokines were detected by ELISA after infection, and the results showed that the expression of IL-6, TNF-α, and IFN-γ levels was higher in virulent strain infection than that of the avirulent group. Results also indicated that the highly virulent strain G. anatis displayed an increased level of adherence. Changes in cytokine profiles in this study suggested that the production of cytokines might influence the microenvironment of oviduct and promote adherence, serving as a possible mechanism inducing cell damage.


Assuntos
Galinhas/microbiologia , Infecções por Pasteurellaceae/veterinária , Pasteurellaceae/patogenicidade , Doenças das Aves Domésticas/microbiologia , Animais , Citocinas/metabolismo , Células Epiteliais/microbiologia , Feminino , Interações Hospedeiro-Patógeno , Oviductos/microbiologia , Óvulo/microbiologia , Infecções por Pasteurellaceae/microbiologia , Virulência
15.
PLoS One ; 11(7): e0158866, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27409077

RESUMO

Strain NI1060 is an oral bacterium responsible for periodontitis in a murine ligature-induced disease model. To better understand its pathogenicity, we have determined the complete sequence of its 2,553,982 bp genome. Although closely related to Pasteurella pneumotropica, a pneumonia-associated rodent commensal based on its 16S rRNA, the NI1060 genomic content suggests that they are different species thriving on different energy sources via alternative metabolic pathways. Genomic and phylogenetic analyses showed that strain NI1060 is distinct from the genera currently described in the family Pasteurellaceae, and is likely to represent a novel species. In addition, we found putative virulence genes involved in lipooligosaccharide synthesis, adhesins and bacteriotoxic proteins. These genes are potentially important for host adaption and for the induction of dysbiosis through bacterial competition and pathogenicity. Importantly, strain NI1060 strongly stimulates Nod1, an innate immune receptor, but is defective in two peptidoglycan recycling genes due to a frameshift mutation. The in-depth analysis of its genome thus provides critical insights for the development of NI1060 as a prime model system for infectious disease.


Assuntos
DNA Bacteriano/genética , Genoma Bacteriano/genética , Pasteurellaceae/classificação , Pasteurellaceae/genética , Periodontite/microbiologia , Sequência de Bases , Lipopolissacarídeos/química , Pasteurellaceae/patogenicidade , Filogenia , RNA Ribossômico 16S/genética , Análise de Sequência de DNA , Fatores de Virulência
16.
Curr Top Microbiol Immunol ; 396: 71-87, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26728064

RESUMO

Histophilus somni resides as part of the normal microflora in the upper respiratory tract of healthy cattle. From this site, the organism can make its way into the lower respiratory tract, where it is one of the important bacterial agents of the respiratory disease complex. If H. somni cells disseminate to the bloodstream, they frequently result in thrombus formation. A series of in vitro investigations have examined potential mechanisms that might contribute to such thrombus formation. Earlier work showed that H. somni can stimulate some bovine endothelial cells to undergo apoptosis. More recent studies indicate that H. somni stimulates endothelial cell tissue factor activity and disrupts intercellular junctions. The net effect is to enhance procoagulant activity on the endothelium surface and to make the endothelial monolayer more permeable to molecules, leukocytes, and perhaps H. somni cells. H. somni also activates bovine platelets, which also can enhance tissue factor activity on the endothelium surface. When exposed to H. somni, bovine neutrophils and mononuclear phagocytes form extracellular traps in vitro. Ongoing research is investigating how the interplay among endothelial cells, platelets, and leukocytes might contribute to the thrombus formation seen in infected cattle.


Assuntos
Interações Hospedeiro-Patógeno , Pasteurellaceae/patogenicidade , Animais , Permeabilidade Capilar , Bovinos , Armadilhas Extracelulares/fisiologia , Imunidade Inata , Trombose/etiologia
17.
Curr Top Microbiol Immunol ; 396: 131-48, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26814887

RESUMO

The lipooligosaccharide (LOS) of Histophilus somni is a multifaceted molecule that provides critical protection to the bacterium against host defenses, may act as an adhesin, and like similar molecules of gram-negative bacteria, is an endotoxin that signals through toll-like receptor 4 and NF-κB to cause inflammation. The lipid A component is responsible for the endotoxic and apoptotic activity of the LOS. The H. somni LOS lacks O-side chains typically characteristic of gram-negative bacteria that have lipopolysaccharide, but has a complex, microheterogeneous outer core. The LOS of disease isolates is capable of undergoing structural and antigenic phase variation of its outer core due to slip-strand mispairing of glycosyltransferase genes that contain repetitive sequences of DNA base pairs. Such variation enables the bacteria to evade bactericidal antibodies made to oligosaccharide antigens. In addition, the LOS can be decorated with phase-variable phosphorylcholine (ChoP), which binds to platelet-activating factor receptor on host cells, thereby aiding in colonization of the upper respiratory tract. However, ChoP is likely not expressed when the bacteria are in systemic sites because ChoP also binds to C-reactive protein, resulting in activation of host complement and promoting bactericidal activity. The structure of some LOS outer core chains is identical to oligosaccharides on host glycosphingolipids of red blood cells, other cells, and merconium (lacto-N-neotetraose, lacto-N-biose, N-acetyllactosamine, etc.). Furthermore, terminal galactose residues on LOS and elsewhere are decorated with sialic acid, which blocks antibody binding, activation of complement, phagocytosis, and intracellular killing. Therefore, antigenic mimicry of host antigens is an important defense mechanism provided by the oligosaccharide component of the LOS to avoid innate and adaptive host defense mechanisms. However, some strains of H. somni isolated from the bovine genital tract, particularly the normal bovine prepuce, are incapable of LOS phase variation, sialylation of the LOS, and expression of ChoP. At least 1 such strain has been shown to be avirulent, underscoring the importance of the LOS as a virulence factor, although this strain is deficient in other factors as well. The structure and arrangement of the inner core glycoses (heptose and 3-deoxy-D-manno-2-octulosnic acid) is remarkably similar to the inner core oligosaccharide on some strains of Neisseria spp., and mutants that contain a truncated LOS oligosaccharide are considerably more serum-sensitive than the parent strain. Therefore, the LOS is a critical component that enables H. somni to resist host defenses and cause disease.


Assuntos
Lipopolissacarídeos/fisiologia , Pasteurellaceae/patogenicidade , Fatores de Virulência/fisiologia , Lipopolissacarídeos/química , Lipopolissacarídeos/isolamento & purificação , Ácido N-Acetilneuramínico/metabolismo , Sialiltransferases/fisiologia
18.
Mol Oral Microbiol ; 31(5): 379-97, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26426681

RESUMO

The QseBC two-component system (TCS) is associated with quorum sensing and functions as a global regulator of virulence. Based on sequence similarity within the sensor domain and conservation of an acidic motif essential for signal recognition, QseBC is primarily distributed in the Enterobacteriaceae and Pasteurellaceae. In Escherichia coli, QseC responds to autoinducer-3 and/or epinephrine/norepinephrine. Binding of epinephrine/norepinephrine is inhibited by adrenergic antagonists; hence QseC functions as a bacterial adrenergic receptor. Aggregatibacter actinomycetemcomitans QseC is activated by a combination of epinephrine/norepinephrine and iron, whereas only iron activates the Haemophilus influenzae sensor. QseC phosphorylates QseB but there is growing evidence that QseB is activated by non-cognate sensors and regulated by dephosphorylation via QseC. Interestingly, the QseBC signaling cascades and regulons differ significantly. In enterohemorrhagic E. coli, QseC induces expression of a second adrenergic TCS and phosphorylates two non-cognate response regulators, each of which induces specific sets of virulence genes. This signaling pathway integrates with other regulatory mechanisms mediated by transcriptional regulators QseA and QseD and a fucose-sensing TCS and likely controls the level and timing of virulence gene expression. In contrast, A. actinomycetemcomitans QseC signals through QseB to regulate genes involved in anaerobic metabolism and energy production, which may prime cellular metabolism for growth in an anaerobic host niche. QseC represents a novel target for therapeutic intervention and small molecule inhibitors already show promise as broad-spectrum antimicrobials. Further characterization of QseBC signaling may identify additional differences in QseBC function and inform further development of new therapeutics to control microbial infections.


Assuntos
Proteínas de Bactérias/metabolismo , Enterobacteriaceae/metabolismo , Proteínas de Escherichia coli/metabolismo , Pasteurellaceae/metabolismo , Receptores Adrenérgicos/metabolismo , Transativadores/metabolismo , Aggregatibacter actinomycetemcomitans/genética , Proteínas de Bactérias/genética , Enterobacteriaceae/genética , Enterobacteriaceae/patogenicidade , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Óperon , Pasteurellaceae/genética , Pasteurellaceae/patogenicidade , Fosforilação , Regiões Promotoras Genéticas , Percepção de Quorum/genética , Receptores Adrenérgicos/genética , Regulon , Transdução de Sinais , Transativadores/genética , Virulência/genética
19.
BMC Res Notes ; 8: 675, 2015 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-26567540

RESUMO

BACKGROUND: Actinobacillus suis disease has been reported in a wide range of vertebrate species, but is most commonly found in swine. A. suis is a commensal of the tonsils of the soft palate of swine, but in the presence of unknown stimuli it can invade the bloodstream, causing septicaemia and sequelae such as meningitis, arthritis, and death. It is genotypically and phenotypically similar to A. pleuropneumoniae, the causative agent of pleuropneumonia, and to other members of the family Pasteurellaceae that colonise tonsils. At present, very little is known about the genes involved in attachment, colonisation, and invasion by A. suis (or related members of the tonsil microbiota). RESULTS: Bioinformatic analyses of the A. suis H91-0380 genome were done using BASys and blastx in GenBank. Forty-seven putative adhesin-associated genes predicted to encode 24 putative adhesins were discovered. Among these are 6 autotransporters, 25 fimbriae-associated genes (encoding 3 adhesins), 12 outer membrane proteins, and 4 additional genes (encoding 3 adhesins). With the exception of 2 autotransporter-encoding genes (aidA and ycgV), both with described roles in virulence in other species, all of the putative adhesin-associated genes had homologues in A. pleuropneumoniae. However, the majority of the closest homologues of the A. suis adhesins are found in A. ureae and A. capsulatus--species not known to infect swine, but both of which can cause systemic infections. CONCLUSIONS: A. suis and A. pleuropneumoniae share many of the same putative adhesins, suggesting that the different diseases, tissue tropism, and host range of these pathogens are due to subtle genetic differences, or perhaps differential expression of virulence factors during infection. However, many of the putative adhesins of A. suis share even greater homology with those of other pathogens within the family Pasteurellaceae. Similar to A. suis, these pathogens (A. capsulatus and A. ureae) cause systemic infections and it is tempting to speculate that they employ similar strategies to invade the host, but more work is needed before that assertion can be made. This work begins to examine adhesin-associated factors that allow some members of the family Pasteurellaceae to invade the bloodstream while others cause a more localised infection.


Assuntos
Actinobacillus suis/genética , Adesinas Bacterianas/genética , Pasteurellaceae/genética , Actinobacillus suis/patogenicidade , Biologia Computacional , Pasteurellaceae/patogenicidade
20.
Avian Dis ; 59(1): 87-93, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26292540

RESUMO

Avibacterium paragallinarum is the causative agent of infectious coryza, an important respiratory disease of chickens. The capsule is an important virulence determinant of many pathogenic bacteria, but the function of the capsule in Av. paragallinarum is not well defined. In this study, acapsular mutants of Av. paragallinarum were constructed by inactivation of the hctA gene using the TargeTron gene knockout system. The acapsular mutants were found to have greater hemagglutination activity than did the wild-type strain. Further, acapsular mutants exhibited an increased ability to adhere to DF-1 cells and to form biofilms on abiotic surfaces. Virulence assays showed that acapsular mutants were less virulent than the wild-type strain. Taken together, these results indicated that loss of capsule increases hemagglutination and adhesion activities but decreases the virulence of Av. paragallinarum. These results could be valuable to further elucidate the function of the capsule and the mechanism of pathogenicity of Av. paragallinarum.


Assuntos
Aderência Bacteriana/fisiologia , Cápsulas Bacterianas/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Pasteurellaceae/metabolismo , Pasteurellaceae/patogenicidade , Animais , Cápsulas Bacterianas/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Pasteurellaceae/genética , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...