Integrated analysis of gut metabolome, microbiome, and exfoliome data in an equine model of intestinal injury

IF 13.8 1区 生物学 Q1 MICROBIOLOGY Microbiome Pub Date : 2024-04-15 DOI:10.1186/s40168-024-01785-1
C. M. Whitfield-Cargile, H. C. Chung, M. C. Coleman, N. D. Cohen, A. M. Chamoun-Emanuelli, I. Ivanov, J. S. Goldsby, L. A. Davidson, I. Gaynanova, Y. Ni, R. S. Chapkin
{"title":"Integrated analysis of gut metabolome, microbiome, and exfoliome data in an equine model of intestinal injury","authors":"C. M. Whitfield-Cargile, H. C. Chung, M. C. Coleman, N. D. Cohen, A. M. Chamoun-Emanuelli, I. Ivanov, J. S. Goldsby, L. A. Davidson, I. Gaynanova, Y. Ni, R. S. Chapkin","doi":"10.1186/s40168-024-01785-1","DOIUrl":null,"url":null,"abstract":"The equine gastrointestinal (GI) microbiome has been described in the context of various diseases. The observed changes, however, have not been linked to host function and therefore it remains unclear how specific changes in the microbiome alter cellular and molecular pathways within the GI tract. Further, non-invasive techniques to examine the host gene expression profile of the GI mucosa have been described in horses but not evaluated in response to interventions. Therefore, the objectives of our study were to (1) profile gene expression and metabolomic changes in an equine model of non-steroidal anti-inflammatory drug (NSAID)-induced intestinal inflammation and (2) apply computational data integration methods to examine host-microbiota interactions. Twenty horses were randomly assigned to 1 of 2 groups (n = 10): control (placebo paste) or NSAID (phenylbutazone 4.4 mg/kg orally once daily for 9 days). Fecal samples were collected on days 0 and 10 and analyzed with respect to microbiota (16S rDNA gene sequencing), metabolomic (untargeted metabolites), and host exfoliated cell transcriptomic (exfoliome) changes. Data were analyzed and integrated using a variety of computational techniques, and underlying regulatory mechanisms were inferred from features that were commonly identified by all computational approaches. Phenylbutazone induced alterations in the microbiota, metabolome, and host transcriptome. Data integration identified correlation of specific bacterial genera with expression of several genes and metabolites that were linked to oxidative stress. Concomitant microbiota and metabolite changes resulted in the initiation of endoplasmic reticulum stress and unfolded protein response within the intestinal mucosa. Results of integrative analysis identified an important role for oxidative stress, and subsequent cell signaling responses, in a large animal model of GI inflammation. The computational approaches for combining non-invasive platforms for unbiased assessment of host GI responses (e.g., exfoliomics) with metabolomic and microbiota changes have broad application for the field of gastroenterology. ","PeriodicalId":18447,"journal":{"name":"Microbiome","volume":null,"pages":null},"PeriodicalIF":13.8000,"publicationDate":"2024-04-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Microbiome","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1186/s40168-024-01785-1","RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MICROBIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

The equine gastrointestinal (GI) microbiome has been described in the context of various diseases. The observed changes, however, have not been linked to host function and therefore it remains unclear how specific changes in the microbiome alter cellular and molecular pathways within the GI tract. Further, non-invasive techniques to examine the host gene expression profile of the GI mucosa have been described in horses but not evaluated in response to interventions. Therefore, the objectives of our study were to (1) profile gene expression and metabolomic changes in an equine model of non-steroidal anti-inflammatory drug (NSAID)-induced intestinal inflammation and (2) apply computational data integration methods to examine host-microbiota interactions. Twenty horses were randomly assigned to 1 of 2 groups (n = 10): control (placebo paste) or NSAID (phenylbutazone 4.4 mg/kg orally once daily for 9 days). Fecal samples were collected on days 0 and 10 and analyzed with respect to microbiota (16S rDNA gene sequencing), metabolomic (untargeted metabolites), and host exfoliated cell transcriptomic (exfoliome) changes. Data were analyzed and integrated using a variety of computational techniques, and underlying regulatory mechanisms were inferred from features that were commonly identified by all computational approaches. Phenylbutazone induced alterations in the microbiota, metabolome, and host transcriptome. Data integration identified correlation of specific bacterial genera with expression of several genes and metabolites that were linked to oxidative stress. Concomitant microbiota and metabolite changes resulted in the initiation of endoplasmic reticulum stress and unfolded protein response within the intestinal mucosa. Results of integrative analysis identified an important role for oxidative stress, and subsequent cell signaling responses, in a large animal model of GI inflammation. The computational approaches for combining non-invasive platforms for unbiased assessment of host GI responses (e.g., exfoliomics) with metabolomic and microbiota changes have broad application for the field of gastroenterology.
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
综合分析马肠道损伤模型中的肠道代谢组、微生物组和脱落组数据
马的胃肠道(GI)微生物群在各种疾病中都有描述。但是,观察到的变化并没有与宿主功能联系起来,因此微生物群的具体变化如何改变胃肠道内的细胞和分子通路仍不清楚。此外,在马匹身上已经出现了检查消化道粘膜宿主基因表达谱的非侵入性技术,但没有针对干预措施进行评估。因此,我们的研究目标是:(1) 在非甾体抗炎药(NSAID)诱发肠道炎症的马模型中分析基因表达和代谢组变化;(2) 应用计算数据整合方法检查宿主与微生物群的相互作用。20 匹马被随机分配到两组中的一组(n = 10):对照组(安慰剂糊剂)或非甾体抗炎药组(苯丁拉宗 4.4 毫克/千克,每天口服一次,连续 9 天)。第 0 天和第 10 天收集粪便样本,分析微生物群(16S rDNA 基因测序)、代谢组(非靶向代谢物)和宿主脱落细胞转录组(脱落组)的变化。利用各种计算技术对数据进行了分析和整合,并从所有计算方法共同确定的特征中推断出潜在的调控机制。苯基丁氮酮诱导了微生物群、代谢组和宿主转录组的改变。数据整合确定了特定细菌属与若干与氧化应激有关的基因和代谢物表达的相关性。微生物群和代谢物的同时变化导致肠粘膜内质网应激和未折叠蛋白反应的启动。综合分析的结果确定了氧化应激和随后的细胞信号反应在大型动物消化道炎症模型中的重要作用。将用于无偏见评估宿主胃肠道反应的非侵入性平台(如脱落细胞组学)与代谢组学和微生物群变化相结合的计算方法在胃肠病学领域具有广泛的应用前景。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
Microbiome
Microbiome MICROBIOLOGY-
CiteScore
21.90
自引率
2.60%
发文量
198
审稿时长
4 weeks
期刊介绍: Microbiome is a journal that focuses on studies of microbiomes in humans, animals, plants, and the environment. It covers both natural and manipulated microbiomes, such as those in agriculture. The journal is interested in research that uses meta-omics approaches or novel bioinformatics tools and emphasizes the community/host interaction and structure-function relationship within the microbiome. Studies that go beyond descriptive omics surveys and include experimental or theoretical approaches will be considered for publication. The journal also encourages research that establishes cause and effect relationships and supports proposed microbiome functions. However, studies of individual microbial isolates/species without exploring their impact on the host or the complex microbiome structures and functions will not be considered for publication. Microbiome is indexed in BIOSIS, Current Contents, DOAJ, Embase, MEDLINE, PubMed, PubMed Central, and Science Citations Index Expanded.
期刊最新文献
Microbial colonisation rewires the composition and content of poplar root exudates, root and shoot metabolomes Succession of rumen microbiota and metabolites across different reproductive periods in different sheep breeds and their impact on the growth and development of offspring lambs Predicting how varying moisture conditions impact the microbiome of dust collected from the International Space Station Microbial mechanisms for higher hydrogen production in anaerobic digestion at constant temperature versus gradient heating Enrichment of novel entomopathogenic Pseudomonas species enhances willow resistance to leaf beetles
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1