当前位置: X-MOL 学术Pediatr. Res. › 论文详情
Our official English website, www.x-mol.net, welcomes your feedback! (Note: you will need to create a separate account there.)
NEC-like intestinal injury is ameliorated by Lactobacillus rhamnosus GG in parallel with SIGIRR and A20 induction in neonatal mice
Pediatric Research ( IF 3.6 ) Pub Date : 2020-02-13 , DOI: 10.1038/s41390-020-0797-6
Alain Cuna 1, 2 , Wei Yu 2 , Heather L Menden 2 , Linda Feng 2 , Pugazhendhi Srinivasan 3 , Susana Chavez-Bueno 1, 2 , Ishfaq Ahmed 3 , Shahid Umar 3 , Venkatesh Sampath 1, 2
Affiliation  

Exaggerated Toll-like receptor (TLR) signaling and intestinal dysbiosis are key contributors to necrotizing enterocolitis (NEC). Lactobacillus rhamnosus GG (LGG) decreases NEC in preterm infants, but underlying mechanisms of protection remain poorly understood. We hypothesized that LGG alleviates dysbiosis and upregulates TLR inhibitors to protect against TLR-mediated gut injury. Effects of LGG (low- and high-dose) on intestinal pro-inflammatory TLR signaling and injury in neonatal mice subjected to formula feeding (FF) and NEC were determined. 16S sequencing of stool and expression of anti-TLR mediators SIGIRR (single immunoglobulin interleukin-1-related receptor) and A20 were analyzed. FF induced mild intestinal injury with increased expression of interleukin-1β (IL-1β) and Kupffer cell (KC) (mouse homolog of IL-8) compared to controls. LGG decreased IL-1β and KC in association with attenuated TLR signaling and increased SIGIRR and A20 expression in a dose-dependent manner. Low- and high-dose LGG had varying effects on gut microbiome despite both doses providing gut protection. Subsequent experiments of LGG on NEC revealed that pro-inflammatory TLR signaling and intestinal injury were also decreased, and SIGIRR and A20 expression increased, in a dose-dependent manner with LGG pre-treatment. LGG protects against intestinal TLR-mediated injury by upregulating TLR inhibitors without major changes in gut microbiome composition.

中文翻译:

在新生小鼠中,鼠李糖乳杆菌 GG 与 SIGIRR 和 A20 诱导同时改善 NEC 样肠道损伤

过度的 Toll 样受体 (TLR) 信号传导和肠道菌群失调是导致坏死性小肠结肠炎 (NEC) 的关键因素。鼠李糖乳杆菌 GG (LGG) 可降低早产儿的 NEC,但其潜在的保护机制仍知之甚少。我们假设 LGG 可缓解生态失调并上调 TLR 抑制剂以防止 TLR 介导的肠道损伤。确定了 LGG(低剂量和高剂量)对接受配方喂养 (FF) 和 NEC 的新生小鼠肠道促炎性 TLR 信号传导和损伤的影响。分析了粪便的 16S 测序和抗 TLR 介质 SIGIRR(单一免疫球蛋白白细胞介素 1 相关受体)和 A20 的表达。与对照相比,FF 诱导轻度肠道损伤,白细胞介素-1β (IL-1β) 和库普弗细胞 (KC)(IL-8 的小鼠同源物)的表达增加。LGG 降低 IL-1β 和 KC 与减弱的 TLR 信号传导相关,并以剂量​​依赖性方式增加 SIGIRR 和 A20 的表达。尽管两种剂量都能提供肠道保护,但低剂量和高剂量 LGG 对肠道微生物组的影响各不相同。LGG 在 NEC 上的后续实验表明,LGG 预处理的促炎性 TLR 信号传导和肠道损伤也减少,SIGIRR 和 A20 表达增加,呈剂量依赖性。LGG 通过上调 TLR 抑制剂来防止肠道 TLR 介导的损伤,而肠道微生物组的组成不会发生重大变化。LGG 在 NEC 上的后续实验表明,LGG 预处理的促炎性 TLR 信号传导和肠道损伤也减少,SIGIRR 和 A20 表达增加,呈剂量依赖性。LGG 通过上调 TLR 抑制剂来防止肠道 TLR 介导的损伤,而肠道微生物组的组成不会发生重大变化。LGG 在 NEC 上的后续实验表明,LGG 预处理的促炎性 TLR 信号传导和肠道损伤也减少,SIGIRR 和 A20 表达增加,呈剂量依赖性。LGG 通过上调 TLR 抑制剂来防止肠道 TLR 介导的损伤,而肠道微生物组的组成不会发生重大变化。
更新日期:2020-02-13
down
wechat
bug