当前位置: X-MOL 学术PLOS Negl. Trop. Dis. › 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.)
Characterisation of novel functionality within the Blastocystis tryptophanase gene.
PLOS Neglected Tropical Diseases ( IF 3.4 ) Pub Date : 2021-09-07 , DOI: 10.1371/journal.pntd.0009730
Steven Santino Leonardi 1 , Feng-Jun Li 2 , Melissa Su-Juan Chee 3 , John Anthony Yason 4 , Hui Yi Tay 3 , John Yu-Shen Chen 3 , Eileen Yiling Koh 1 , Cynthia Ying-Xin He 2 , Kevin Shyong-Wei Tan 1
Affiliation  

In recent years, the human gut microbiome has been recognised to play a pivotal role in the health of the host. Intestinal homeostasis relies on this intricate and complex relationship between the gut microbiota and the human host. While much effort and attention has been placed on the characterization of the organisms that inhabit the gut microbiome, the complex molecular cross-talk between the microbiota could also exert an effect on gastrointestinal conditions. Blastocystis is a single-cell eukaryotic parasite of emerging interest, as its beneficial or pathogenic role in the microbiota has been a subject of contention even to-date. In this study, we assessed the function of the Blastocystis tryptophanase gene (BhTnaA), which was acquired by horizontal gene transfer and likely to be of bacterial origin within Blastocystis. Bioinformatic analysis and phylogenetic reconstruction revealed distinct divergence of BhTnaA versus known bacterial homologs. Despite sharing high homology with the E. coli tryptophanase gene, we show that Blastocystis does not readily convert tryptophan into indole. Instead, BhTnaA preferentially catalyzes the conversion of indole to tryptophan. We also show a direct link between E. coli and Blastocystis tryptophan metabolism: In the presence of E. coli, Blastocystis ST7 is less able to metabolise indole to tryptophan. This study examines the potential for functional variation in horizontally-acquired genes relative to their canonical counterparts, and identifies Blastocystis as a possible producer of tryptophan within the gut.

中文翻译:

囊胚色氨酸酶基因内新功能的表征。

近年来,人类肠道微生物群已被认为在宿主的健康中起着举足轻重的作用。肠道稳态依赖于肠道微生物群与人类宿主之间错综复杂的关系。尽管对居住在肠道微生物群中的生物体的特征进行了大量的努力和关注,但微生物群之间复杂的分子串扰也可能对胃肠道状况产生影响。囊胚是一种新出现的单细胞真核寄生虫,因为它在微生物群中的有益或致病作用至今仍是争论的主题。在这项研究中,我们评估了囊胚色氨酸酶基因 (BhTnaA) 的功能,该基因是通过水平基因转移获得的,可能来源于囊胚内的细菌。生物信息学分析和系统发育重建揭示了 BhTnaA 与已知细菌同源物的明显分歧。尽管与大肠杆菌色氨酸酶基因具有高度同源性,但我们表明囊胚不会轻易将色氨酸转化为吲哚。相反,BhTnaA 优先催化吲哚向色氨酸的转化。我们还展示了大肠杆菌和囊胚的色氨酸代谢之间的直接联系:在大肠杆菌存在的情况下,囊胚 ST7 将吲哚代谢为色氨酸的能力较低。这项研究检查了水平获得基因相对于其典型对应物的功能变异的可能性,并确定囊胚是肠道内色氨酸的可能生产者。尽管与大肠杆菌色氨酸酶基因具有高度同源性,但我们表明囊胚不会轻易将色氨酸转化为吲哚。相反,BhTnaA 优先催化吲哚向色氨酸的转化。我们还展示了大肠杆菌和囊胚的色氨酸代谢之间的直接联系:在大肠杆菌存在的情况下,囊胚 ST7 将吲哚代谢为色氨酸的能力较低。这项研究检查了水平获得基因相对于其典型对应物的功能变异的可能性,并确定囊胚是肠道内色氨酸的可能生产者。尽管与大肠杆菌色氨酸酶基因具有高度同源性,但我们表明囊胚不会轻易将色氨酸转化为吲哚。相反,BhTnaA 优先催化吲哚向色氨酸的转化。我们还展示了大肠杆菌和囊胚的色氨酸代谢之间的直接联系:在大肠杆菌存在的情况下,囊胚 ST7 将吲哚代谢为色氨酸的能力较低。这项研究检查了水平获得基因相对于其典型对应物的功能变异的可能性,并确定囊胚是肠道内色氨酸的可能生产者。大肠杆菌和囊胚的色氨酸代谢:在大肠杆菌存在的情况下,囊胚 ST7 将吲哚代谢为色氨酸的能力较低。这项研究检查了水平获得基因相对于其典型对应物的功能变异的可能性,并确定囊胚是肠道内色氨酸的可能生产者。大肠杆菌和囊胚的色氨酸代谢:在大肠杆菌存在的情况下,囊胚 ST7 将吲哚代谢为色氨酸的能力较低。这项研究检查了水平获得基因相对于其典型对应物的功能变异的可能性,并确定囊胚是肠道内色氨酸的可能生产者。
更新日期:2021-09-07
down
wechat
bug