Salmonella Infantis

  • 文章类型: Journal Article
    Dairy cows can be reservoirs of foodborne pathogens such as Salmonella that pose serious public health risks to humans. The study was designed to examine the molecular epidemiology and antimicrobial resistance profiles of Salmonella isolates from dairy heifer calves and adult lactating cows in the pasture-based system of Australia. A total of 838 animals (328 heifer calves and 510 lactating cows) from 22 farms were sampled. Overall, 54 Salmonella isolates were recovered (calves 28/328 and cows 26/510). A herd-level Salmonella prevalence of 50% (95% confidence interval: 31%-69%) was recorded. Within-herd prevalence for Salmonella ranged between 4%-29% and 4%-45% among the heifer calves and adult lactating cows, respectively. Three different serovars were identified with Salmonella Infantis being the most common serovar (n = 33, 61%) followed by Salmonella Kiambu (n = 20, 37.0%) and one isolate of Salmonella Cerro (2%). The highest antimicrobial resistance prevalence of Salmonella isolates was found against streptomycin (n = 31, 57%), followed by cefoxitin (n = 12, 22%), ceftriaxone (n = 2, 4%), and chloramphenicol (n = 1, 2%). Multiple class resistance was observed on 4 isolates against cefoxitin, chloramphenicol, and streptomycin. Multilocus sequence types ST32 (61%), ST309 (37%), and ST367 (2%) were strongly linked to the serovars Salmonella Infantis, Salmonella Kiambu, and Salmonella Cerro, respectively. Whole genome sequencing of Salmonella isolates detected only 2 resistance genes: aac(6\') gene that confers resistance against aminoglycosides among 40.7% of the isolates, and a single isolate positive for the blaDHA-16 gene. Two distinct clusters among the serovars were observed suggesting 2 independent sources of spread. Despite the low prevalence of antimicrobial resistance among Salmonella from the dairy farms, our findings contribute to the regional and national understanding of antimicrobial resistance in dairy herds in Australia. There is need for continued antimicrobial resistance stewardship and surveillance programs to ensure the production of high-quality food products and the long-term protection of both animal and human health.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    近年来,婴儿沙门氏菌已成为世界范围内引起胃肠炎的主要临床病原体。作为细胞内病原体,沙门氏菌已经进化为操纵和受益于细胞死亡信号通路。在这项研究中,我们发现婴儿链球菌通过磷酸化Akt抑制感染的Caco-2细胞的凋亡。值得注意的是,Akt磷酸化以不连续的方式观察:入侵后立即0.5小时,然后在峰细胞溶质复制之前。单细胞分析显示,第二阶段仅在3-4hpi时由胞质过度复制的细菌诱导。接下来,发现Akt介导的凋亡抑制是由沙门氏菌SopB引发的。此外,Akt磷酸化增加了Bcl-2的线粒体定位,以防止Bax在线粒体膜上的寡聚化,维持线粒体网络稳态以抵抗细胞凋亡。此外,美国婴儿诱导的焦亡,如增加的caspase-1(p10)和GSDMS-N水平证明。相比之下,感染ΔSopB菌株的细胞表现出更快但不那么严重的焦亡,并且细菌负荷较少。结果表明,婴儿链球菌SopB介导的Akt磷酸化延迟了细胞凋亡,但加重了它的严重程度。野生型菌株在小鼠中也比ΔSopB菌株引起更严重的腹泻和肠道炎症损伤。这些发现表明,婴儿链球菌通过间歇性激活Akt来延迟细胞死亡,允许足够的时间进行复制,从而引起更严重的炎症。
    Salmonella Infantis has emerged as a major clinical pathogen causing gastroenteritis worldwide in recent years. As an intracellular pathogen, Salmonella has evolved to manipulate and benefit from the cell death signaling pathway. In this study, we discovered that S. Infantis inhibited apoptosis of infected Caco-2 cells by phosphorylating Akt. Notably, Akt phosphorylation was observed in a discontinuous manner: immediately 0.5 h after the invasion, then before peak cytosolic replication. Single-cell analysis revealed that the second phase was only induced by cytosolic hyper-replicating bacteria at 3-4 hpi. Next, Akt-mediated apoptosis inhibition was found to be initiated by Salmonella SopB. Furthermore, Akt phosphorylation increased mitochondrial localization of Bcl-2 to prevent Bax oligomerization on the mitochondrial membrane, maintaining the mitochondrial network homeostasis to resist apoptosis. In addition, S. Infantis induced pyroptosis, as evidenced by increased caspase-1 (p10) and GSDMS-N levels. In contrast, cells infected with the ΔSopB strain displayed faster but less severe pyroptosis and had less bacterial load. The results indicated that S. Infantis SopB-mediated Akt phosphorylation delayed pyroptosis, but aggravated its severity. The wild-type strain also caused more severe diarrhea and intestinal inflammatory damage than the ΔSopB strain in mice. These findings revealed that S. Infantis delayed the cells\' death by intermittent activation of Akt, allowing sufficient time for replication, thereby causing more severe inflammation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    沙门氏菌既是猪临床疾病的原因,也是食源性疾病向人类传播的来源。乳酸杆菌和芽孢杆菌通常用作抗生素替代品以防止沙门氏菌感染。在这项研究中,在猪模型中,我们评估了约氏乳杆菌L531,地衣芽孢杆菌BL1721和枯草芽孢杆菌BS1715(LBB-mix)的特定混合物预防感染肠道沙门氏菌的效果.在S.Infantis攻击之前,将LBB混合物口服给予新断奶的仔猪七天。LBB混合预处理改善了婴儿链球菌引起的发烧,白细胞增多,增长业绩损失,和回肠炎症。LBB混合物的预先给药减少了粪便中沙门氏菌的数量,但增加了回肠中杯状细胞的数量。美国婴儿感染导致回肠细胞死亡增加,LBB混合消费减弱了这种增加。在用S.Infantis攻击的猪的回肠中,Claudin1和裂解的caspase-1表达降低,但不是用LBB-mix预处理的猪。总之,我们的数据表明,选定的LBB-mix通过减轻炎症和维持猪的肠粘膜屏障完整性,对控制婴儿链球菌感染具有积极作用。
    Salmonella is important as both a cause of clinical disease in swine and as a source of food-borne transmission of disease to humans. Lactobacillus and Bacillus are often used as antibiotic substitutes to prevent Salmonella infection. In this study, we evaluated the effects of a select mixture of Lactobacillus johnsonii L531, Bacillus licheniformis BL1721 and Bacillus subtilis BS1715 (LBB-mix) in prevention of Salmonella enterica serovar Infantis infection in a pig model. LBB-mix was orally administered to newly weaned piglets for seven days before S. Infantis challenge. LBB-mix pretreatment ameliorated S. Infantis-induced fever, leukocytosis, growth performance loss, and ileal inflammation. Pre-administration of LBB-mix reduced the number of Salmonella in the feces but increased the number of goblet cells in the ileum. S. Infantis infection resulted in an increase in cell death in the ileum, this increase was attenuated by LBB-mix consumption. Claudin 1 and cleaved caspase-1 expression was decreased in the ileum of pigs challenged with S. Infantis, but not in pigs pretreated with LBB-mix. In conclusion, our data indicate that a select LBB-mix has positive effects on controlling S. Infantis infection via alleviating inflammation and maintaining the intestinal mucosal barrier integrity in pigs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    In the current study, we screened Lactobacillus strains isolated from the colon of clinically healthy weaned piglets for potential probiotic properties and isolated Lactobacillus. johnsonii L531, which produced high levels of beneficial metabolites (butyric, acetic, and lactic acid) in vitro. We also evaluated the efficacy of this metabolites-producing probiotic in treating Salmonella. Infantis infection. Oral administration of L. johnsonii L531 to newly weaned piglets significantly decreased levels of Salmonella colonization in colonic and jejunal contents, accelerated the clearance of Salmonella in feces after infection, and reduced S. Infantis translocation to the spleen. Pretreatment with SCFAs-promoting probiotic L. johnsonii L531 significantly ameliorated the depletion of SCFAs induced by S. Infantis infection and led to significantly greater weight gain and better feed conversion ratios compared to piglets challenged only with S. Infantis. These data provide further evidence that SCFAs-promoting probiotic L. johnsonii L531 treatment could be a suitable nonantibiotic alternative for controlling Salmonella infection and maintaining metabolic homeostasis, thereby enhancing the gut health of piglets during the critical weaning period.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Salmonella enterica serovar Infantis (S. Infantis) is a common source of foodborne gastroenteritis worldwide. Here, Lactobacillus rhamnosus GG (LGG) was administrated to weaned piglets for 1 week before S. Infantis challenge. S. Infantis caused decreased ileal mucosal microbiota diversity, a dramatic Lactobacillus amylovorus bloom, and decreased abundance of Arsenicicoccus, Janibacter, Kocuria, Nocardioides, Devosia, Paracoccus, Psychrobacter, and Weissella. The beneficial effect of LGG correlated with the moderate expansion of L. amylovorus, L. agilis, and several members of the phyla Proteobacteria, Firmicutes, and Bacteroidetes. S. Infantis translocation to the liver was decreased in the LGG-pretreated piglets. An in vitro model of LGG and S. Infantis co-incubation (involving the porcine intestinal epithelial cell line IPEC-J2) was established, and nalidixic acid was used to kill the extracellular S. Infantis. LGG suppressed the initial S. Infantis invasion in the IPEC-J2 cells and deceased the rate of cell death. LGG inhibited S. Infantis-induced autophagy and promoted epidermal growth factor receptor (EGFR) and Akt phosphorylation in both the ileum and IPEC-J2 cells. Our findings suggest that LGG inhibited S. Infantis-induced autophagy by promoting EGFR-mediated activation of the negative mediator Akt, which, in turn, suppressed intestinal epithelial cell death and thus restricted systemic S. Infantis infection. LGG can restore the gut microbiota balance and preserve the autophagy-related intestinal epithelial barrier, thereby controlling infections.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    肠沙门氏菌的高发率婴儿(S.婴儿)感染对非伤寒沙门氏菌胃肠炎的发展构成重大风险。然而,预防或治疗感染的有效策略仍然难以捉摸。这里,我们探讨了益生菌鼠李糖乳杆菌GG(LGG)在猪模型中预防婴儿链球菌感染的作用。在S.Infantis攻击之前,将益生菌LGG(1.0×1010CFU/天)口服给予新断奶仔猪1周。LGG预处理降低了腹泻的严重程度并减轻了由S.Infantis引起的肠道炎症。LGG的预施用将沙门氏菌从空肠粘膜的定植中排除,但增加了粪便中双歧杆菌的丰度。LGG促进CD4+T-bet+IFNγ+T细胞的扩增,但减弱S.Infantis攻击后外周血中CD4+IFNγ+T细胞百分比和血清白细胞介素(IL)-22水平的增加。在小肠里,LGG预处理上调转录因子T-bet的表达,但下调婴儿链球菌诱导的Peyer斑块中CD4+IFNγ+T细胞的增加和空肠中IL-7Rα的表达。值得注意的是,LGG处理的猪响应于婴儿链球菌感染而在回肠中具有增强的IL-22表达和激活的STAT3。用LGG预处理猪还提高了肠IL-22结合蛋白的产生,以响应婴儿链球菌攻击。相比之下,LGG消耗减少了空肠中婴儿链球菌诱导的表达CCL20的细胞数量的增加。我们的结果表明,LGG改善由S.Infantis引起的肠道炎症的机制涉及T-bet的上调,STAT3的激活和CCL20的下调。
    The high rate of Salmonella enterica serovar Infantis (S. Infantis) infection poses significant risk for the development of non-typhoidal Salmonella gastroenteritis. However, efficient strategies to prevent or treat the infection remain elusive. Here, we explored the effect of the probiotic Lactobacillus rhamnosus GG (LGG) administration in preventing S. Infantis infection in a pig model. Probiotic LGG (1.0 × 1010 CFU/day) was orally administered to newly weaned piglets for 1 week before S. Infantis challenge. LGG pretreatment reduced the severity of diarrhea and alleviated intestinal inflammation caused by S. Infantis. Pre-administration of LGG excluded Salmonella from colonization of the jejunal mucosa but increased the abundance of Bifidobacterium in the feces. LGG promoted the expansion of CD4+ T-bet+ IFNγ+ T cells but attenuated S. Infantis-induced increases in the percentage of CD4+ IFNγ+ T cells and serum interleukin (IL)-22 levels in peripheral blood after S. Infantis challenge. In the small intestine, LGG pretreatment upregulated expression of the transcription factor T-bet but downregulated the S. Infantis-induced increase of CD4+ IFNγ+ T cells in Peyer\'s patches and IL-7Rα expression in the jejunum. Notably, LGG-treated pigs had enhanced expression of IL-22 and activated STAT3 in the ileum in response to S. Infantis infection. Pretreatment of pigs with LGG also elevated intestinal IL-22-binding protein production in response to S. Infantis challenge. In contrast, LGG consumption reduced the S. Infantis-induced increase in the number of CCL20-expressing cells in the jejunum. Our results suggest that the mechanism by which LGG ameliorates the intestinal inflammation caused by S. Infantis involves the upregulation of T-bet, activation of STAT3, and downregulation of CCL20.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号